US20030199463A1 - DNA enzyme to inhibit plasminogen activator inhibitor-1 - Google Patents

DNA enzyme to inhibit plasminogen activator inhibitor-1 Download PDF

Info

Publication number
US20030199463A1
US20030199463A1 US10/128,706 US12870602A US2003199463A1 US 20030199463 A1 US20030199463 A1 US 20030199463A1 US 12870602 A US12870602 A US 12870602A US 2003199463 A1 US2003199463 A1 US 2003199463A1
Authority
US
United States
Prior art keywords
pai
subject
nucleotides
nucleic acid
catalytic nucleic
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
US10/128,706
Inventor
Silviu Itescu
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.)
Individual
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 US10/128,706 priority Critical patent/US20030199463A1/en
Priority to JP2003587981A priority patent/JP4574992B2/en
Priority to DE60333747T priority patent/DE60333747D1/en
Priority to PCT/US2003/012767 priority patent/WO2003091456A1/en
Priority to CN038147149A priority patent/CN1662663B/en
Priority to US10/512,496 priority patent/US7662794B2/en
Priority to AT03724216T priority patent/ATE477324T1/en
Priority to ZA200408776A priority patent/ZA200408776B/en
Priority to AU2003231089A priority patent/AU2003231089B2/en
Priority to CA2483007A priority patent/CA2483007C/en
Priority to EP03724216A priority patent/EP1501948B1/en
Publication of US20030199463A1 publication Critical patent/US20030199463A1/en
Priority to IL164674A priority patent/IL164674A/en
Priority to AU2009201027A priority patent/AU2009201027B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • C07K14/8121Serpins
    • C07K14/8132Plasminogen activator inhibitors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/12Type of nucleic acid catalytic nucleic acids, e.g. ribozymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates

Definitions

  • plasmin which is derived from plasminogen through activation by urokinase-type plasminogen activator (u-PA) expressed on the surface of the infiltrating bone-marrow derived cells (1-3). More recently, it has been suggested that the role of cell surface u-PA may be to facilitate exposure of cryptic cell attachment sites necessary for cell migration (4).
  • PAI-1 plasminogen activator inhibitor-1
  • u-PA plasminogen activator inhibitor-1
  • u-PA plasminogen activator inhibitor-1
  • a proteinase such as u-PA
  • u-PA plasminogen activator inhibitor-1
  • Removal of PAI-1 from vitronectin exposes the epitope on vitronectin necessary for binding to another of its ligands, the integrin alpha v beta 3 (4,7).
  • Antisense oligonucleotides hybridize with their complementary target site in mRNA, blocking translation to protein by sterically inhibiting ribosome movement or by triggering cleavage by endogenous RNAse H (10). Although current constructs are made more resistant to degradation by serum through phosphorothioate linkages, non-specific biological effects due to “irrelevant cleavage” of non-targeted mRNA remains a major concern (11).
  • Ribozymes are naturally-occurring RNA molecules that contain catalytic sites, making them more potent agents than antisense oligonucleotides.
  • wider use of ribozymes has been hampered by their susceptibility to chemical and enzymatic degradation and restricted target site specificity (12).
  • RNA molecules with catalytic activity for specific RNA sequences (13-16). These DNA enzymes exhibit greater catalytic efficiency than hammerhead ribozymes, producing a rate enhancement of approximately 10 million-fold over the spontaneous rate of RNA cleavage, offer greater substrate specificity, are more resistant to chemical and enzymatic degradation, and are far cheaper to synthesize.
  • This invention provides a catalytic nucleic acid that specifically cleaves an mRNA encoding a Plasminogen Activator Inhibitor-1 (PAI-1) comprising, in 5′ to 3′ order:
  • sequence of the nucleotides in each binding domain is complementary to a sequence of ribonucleotides in the PAI-1-encoding mRNA and wherein the catalytic nucleic acid hybridizes to and specifically cleaves the PAI-1-encoding mRNA.
  • This invention further provides a pharmaceutical composition comprising the instant catalytic nucleic acid and a pharmaceutically acceptable carrier.
  • This invention further provides method of specifically inhibiting the expression of PAI-1 in a cell that would otherwise express PAI-1, comprising contacting the cell with the instant catalytic nucleic acid so as to specifically inhibit the expression of PAI-1 in the cell.
  • This invention further provides a method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the instant catalytic nucleic acid effective to specifically inhibit the expression of PAI-1 in the subject's cells.
  • This invention further provides a method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the instant pharmaceutical composition effective to specifically inhibit the expression of PAI-1 in the subject's cells.
  • This invention further provides a method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the instant pharmaceutical composition effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease.
  • This invention further provides a method of treating a fibrotic disease in a subject involving fibrogenesis which comprises administering to the subject an amount of the instant pharmaceutical composition effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease.
  • This invention further provides an oligonucleotide comprising consecutive nucleotides that hybridizes with a PAI-1-encoding mRNA under conditions of high stringency and is between 8 and 40 nucleotides in length.
  • This invention further provides the instant oligonucleotide, wherein the human PAI-1-encoding mRNA comprises consecutive nucleotides, the sequence of which is set forth in SEQ ID NO:5.
  • This invention further provides a method of treating a subject which comprises administering to the subject an amount of the instant oligonucleotide of effective to inhibit expression of a PAI-1 in the subject so as to thereby treat the subject.
  • This invention further provides a method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease.
  • This invention further provides a method of treating a fibrotic disease in a subject involving fibrogenesis in the subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease.
  • E1 SEQ ID NO:2
  • E3 SEQ ID NO:3
  • This figure also shows the control DNA enzyme E0 (SEQ ID NO:7), an oligonucleotide S1 (SEQ ID NO:8), an oligonucleotide transcript (SEQ ID NO:9) and oligonucleotide S2 (SEQ ID NO:10).
  • E0 SEQ ID NO:7
  • SEQ ID NO:8 an oligonucleotide S1
  • SEQ ID NO:9 oligonucleotide transcript
  • S2 oligonucleotide S2
  • (C) This figure shows E1 also cleaved larger 32P-labeled fragments of human PAI-1 mRNA, prepared by in vitro transcription, in a time- and concentration-dependent manner.
  • (D) This figure shows the sequence-specific nature of the DNA enzymatic cleavage: the control DNA enzyme E0, containing an identical catalytic domain to E1 and E3, but scrambled sequences in the flanking arms, caused no cleavage of human PAI-1 mRNA transcripts.
  • A This figure shows that the DNA enzyme E2 cleaved the 23-base oligonucleotide S2 (SEQ ID NO:10), synthesized from the sequence of rat PAI-1 mRNA, in a dose- and time-dependent manner.
  • B This figure shows that E2 also cleaved a rat PAI-1 mRNA transcript in a dose-dependent manner by 2-4 hours to give the 156 nucleotide cleavage product.
  • C This figure shows the rat S2 PAI-1 mRNA transcript (SEQ ID NO:10) differs by only one nucleotide from the human mRNA PAI-1 transcript (SEQ ID NO:9) which can be cleaved by E3.
  • (B) This figure shows the effect of endothelial cell transfection with E2 DNA enzyme on TGF-beta mediated induction of PAI-1 protein. Endothelial cells transfected with scrambled DNA enzyme demonstrated approximately 50% increase in cytoplasmic PAI-1 E0 protein as detected by Western blot. In contrast, this effect was almost completely abrogated by transfection with the PAI-1 DNA enzyme E2.
  • (C) This figure shows that whereas the DNA enzyme was almost completely degraded within 6 hours of cell culture in medium containing 20% serum, it remained intact during the entire 24 hours of cell culture with medium containing 2% serum.
  • FIGS. 5 A-C (A) This figure shows inhibition of cardiomyocyte apoptosis in the peri-infarct region by E2. (B) This figure shows the percentage recovery in left ventricular ejection fraction (LVEF) after treatment with E2 and after treatment with E2 and angioblasts.
  • LVEF left ventricular ejection fraction
  • FIG. 6 This figure shows the DNA sequence encoding human Plasminogen Activator Inhibitor-1 protein (SEQ ID NO:5).
  • FIG. 7 This figure shows the DNA sequence encoding rat Plasminogen Activator Inhibitor-1 protein (SEQ ID NO:15).
  • FIG. 8 This figure shows the amino acid sequence of human Plasminogen Activator Inhibitor-1 protein (SEQ ID NO:6).
  • FIG. 9 This figure shows the amino acid sequence of rat Plasminogen Activator Inhibitor-1 protein (SEQ ID NO:16).
  • FIG. 10 This figure shows possible 5′-AT-3′ cleavage sites on human PAI-1 mRNA (shown as corresponding DNA, SEQ ID NO:5) for catalytic deoxyribonucleic acids. Bold indicates the protein coding region, capital letters indicate consensus cleavage sites.
  • FIG. 11 This figure shows possible 5′-AC-3′ cleavage sites on human PAI-1 mRNA (shown as corresponding DNA, SEQ ID NO:5) for catalytic deoxyribonucleic acids. Bold indicates the protein coding region, capital letters indicate consensus cleavage sites.
  • FIG. 12 This figure shows possible cleavage sites on human PAI-1 mRNA coding region (shown as corresponding DNA, SEQ ID NO:17) for catalytic hammerhead ribonucleic acids.
  • Uppercase “T” represents cleavage site.
  • administering shall mean any of the various methods and delivery systems known to those skilled in the art.
  • the administering can be performed, for example, via implant, transmucosally, transdermally and subcutaneously, orally, parenterally, topically, by cardiac injection, by inhalation, by catheter e.g. retrograde ureteric catheter, by intra-arterial injection.
  • Catalytic shall mean the functioning of an agent as a catalyst, i.e. an agent that increases the rate of a chemical reaction without itself undergoing a permanent structural change.
  • Consensus sequence shall mean a nucleotide sequence of at least two residues in length between which catalytic nucleic acid cleavage occurs.
  • consensus sequences include for catalytic deoxyribonucleic acids are purine:pyrimidine e.g. “A:U” and “G:U”.
  • “Plasminogen Activator Inhibitor-1 Protein” shall mean the protein encoded by the nucleotide sequence identified as (SEQ ID NO:5) and having the amino acid sequence shown in SEQ ID NO:6, when identified as human and the protein encoded by the nucleotide sequence identified as SEQ ID NO:15 and having the amino acid sequence shown in SEQ ID NO:16 when identified as originating from rat, and any variants of either thereof, whether artificial or naturally occurring. Variants include, without limitation, homologues, post-translational modifications, mutants and polymorphisms.
  • Plasminogen Activator Inhibitor-1 mRNA shall mean a mRNA molecule comprising a sequence which encodes Plasminogen Activator Inhibitor-1 Protein.
  • Plasminogen Activator Inhibitor-1 mRNA includes, without limitation, protein-encoding sequences as well as the 5′ and 3′ non-protein-encoding sequences.
  • Hybridize shall mean the annealing of one single-stranded nucleic acid molecule to another nucleic acid molecule based on sequence complementarity.
  • the propensity for hybridization between nucleic acids depends on the temperature and ionic strength of their milieu, the length of the nucleic acids and the degree of complementarity. The effect of these parameters on hybridization is well known in the art (see 38).
  • “Inhibit” shall mean to slow, stop or otherwise impede.
  • “Stringent conditions” or “Stringency”, shall refer to the conditions for hybridization as defined by the nucleic acid, salt, and temperature. These conditions are well known in the art and may be altered. Numerous equivalent conditions comprising either low or high stringency depend on factors such as the length and nature of the sequence (DNA, RNA, base composition), nature of the target (DNA, RNA, base composition), milieu (in solution or immobilized on a solid substrate), concentration of salts and other components (e.g., formamide, dextran sulfate and/or polyethylene glycol), and temperature of the reactions (within a range from about 5° C. below the melting temperature of the probe to about 20° C. to 25° C. below the melting temperature).
  • salts and other components e.g., formamide, dextran sulfate and/or polyethylene glycol
  • One or more factors be may be varied to generate conditions of either low or high stringency different from, but equivalent to, the above listed conditions.
  • the stringency of hybridization may be altered in order to identify or detect identical or related polynucleotide sequences.
  • Nucleic acid shall include any nucleic acid, including, without limitation, DNA, RNA, oligonucleotides, or polynucleotides, and analogs or derivatives thereof.
  • the nucleotides that form the nucleic acid may be nucleotide analogs or derivatives thereof.
  • the nucleic acid may incorporate non nucleotides.
  • Nucleotides shall include without limitation nucleotides and analogs or derivatives thereof.
  • nucleotides may comprise the bases A, C, G, T and U, as well as derivatives thereof. Derivatives of these bases are well known in the art, and are exemplified in PCR Systems, Reagents and Consumables (Perkin Elmer Catalogue 1996-1997, Roche Molecular Systems, Inc., Branchburg, N.J., USA).
  • “Pharmaceutically acceptable carrier” shall mean any of the various carriers known to those skilled in the art.
  • cleave when referring to the action of one of the instant catalytic deoxyribonucleic acid molecules on a target mRNA molecule, shall mean to cleave the target mRNA molecule without cleaving another mRNA molecule lacking a sequence complementary to either of the catalytic deoxyribonucleic acid molecule's two binding domains.
  • Subject shall mean any animal, such as a human, a primate, a mouse, a rat, a guinea pig or a rabbit.
  • DNA enzyme DNA enzyme
  • DNAzyme DNAzyme
  • catalytic deoxyribonucleic acid catalytic deoxyribonucleic acid
  • Vector shall include, without limitation, a nucleic acid molecule that can be used to stably introduce a specific nucleic acid sequence into the genome of an organism.
  • This invention provides a catalytic nucleic acid that specifically cleaves an mRNA encoding a Plasminogen Activator Inhibitor-1 (PAI-1) comprising, in 5′ to 3′ order:
  • sequence of the nucleotides in each binding domain is complementary to a sequence of ribonucleotides in the PAI-1-encoding mRNA and wherein the catalytic nucleic acid hybridizes to and specifically cleaves the PAI-1-encoding mRNA.
  • the catalytic deoxyribonucleic acid molecules can cleave the PAI-1-encoding mRNA at the linkage between 5′-A (or G) and U(T)-3′ (or C) i.e. at purine:pyrimidine consensus sequences.
  • the catalytic deoxyribonucleic acid molecules cleaves the PAI-1 mRNA at the linkage of any 5′-AU-3′; 5′-AC-3′; 5′-GU-3′; or 5′-GC-3′ site within the mRNA.
  • catalytic nucleic acids can be designed based on the consensus cleavage sites 5′-purine:pyrimidine-3′ in the mRNA sequence (GenBank Accession#: M16006) (36). Those potential cleavage sites located on an open loop of the mRNA according to RNA folding software e.g. RNAdraw 2.1 are particularly preferred as targets (22).
  • the DNA based catalytic nucleic acids can utilize the structure where two sequence-specific arms are attached to a catalytic core (SEQ ID NO:1) based on the PAI-1-encoding mRNA sequence (corresponding DNA shown as SEQ ID NO:5). Further examples of catalytic DNA structure are detailed in (23) and (24).
  • Commercially available mouse brain polyA-RNA (Ambion) can serve as a template in the in vitro cleavage reaction to test the efficiency of the catalytic deoxyribonucleic acids.
  • Catalytic nucleic acid molecules can cleave PAI-1-encoding mRNA at each and any of the consensus sequences therein. Since catalytic ribo- and deoxyribo-nucleic acid consensus sequences are known, and the PAI-1-encoding mRNA sequence is known, one of ordinary skill could readily construct a catalytic ribo- or deoxyribo nucleic acid molecule directed to any of the PAI-1-encoding mRNA consensus sequences based on the instant specification. In preferred embodiments of this invention the catalytic deoxyribonucleic acids include the 10-23 structure.
  • Cleavage of PAI-1 mRNA by DNAzyme may occur at 264 cleavage sites in the coding region of human PAI-1-encoding mRNA, including 51 5′-AT-3′ (-AU-) sites, 76 -AC- sites, 53 -GT- (-GU-) sites and 84 -GC- sites. See FIGS. 10 and 11 for AT and AC cleavage sites respectively, represented on the DNA sequence corresponding to the PAI-1-encoding mRNA.
  • cleavage sites include nucleotide 76, 82, 152, 159, 254, 277, 316, 328, 333, 340, 344, 355, 374, 391, 399, 410, 415, 433, 449, 472, 543, 547, 550, 554, 583, 586, 644, 717, 745, 748, 773, 794, 800, 811, 847, 853, 866, 901, 919, 939, 1027, 1036, 1120, 1125, 1168, 1183, 1198, 1201, 1204, 1261, and 1273 of SEQ ID NO:5.
  • catalytic ribonucleic acids include hairpin and hammerhead ribozymes.
  • the catalytic ribonucleic acid molecule is formed in a hammerhead (25) or hairpin motif (26,27,28), but may also be formed in the motif of a hepatitis delta virus (29), group I intron (35), RNaseP RNA (in association with an RNA guide sequence) (30,31) or Neurospora VS RNA (32,33,34).
  • Hammerhead ribozymes can cleave any 5′-NUH-3′ triplets of a mRNA, where U is conserved and N is any nucleotide and H can be C,U,A, but not G. For example, there are 151 sites which can be cleaved by a hammerhead ribozyme in human PAI-1-encoding mRNA coding region. See FIG. 12.
  • Cleaving of PAI-1-encoding mRNA with catalytic nucleic acids interferes with one or more of the normal functions of PAI-1-encoding mRNA.
  • the functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA.
  • nucleotides of the first binding domain comprise at least one deoxyribonucleotide. In another embodiment the nucleotides of the second binding domain comprise at least one deoxyribonucleotide. In one embodiment the nucleotides of the first binding domain comprise at least one deoxyribonucleotide derivative. In another embodiment the nucleotides of the second binding domain comprise at least one deoxyribonucleotide derivative. In one embodiment the nucleotides of the first binding domain comprise at least one ribonucleotide. In another embodiment the nucleotides of the second binding domain comprise at least one ribonucleotide.
  • nucleotides of the first binding domain comprise at least one ribonucleotide derivative. In another embodiment the nucleotides of the second binding domain comprise at least one ribonucleotide derivative. In one embodiment the nucleotides of the first binding domain comprise at least one modified base. In another embodiment the nucleotides of the second binding domain comprise at least one modified base.
  • the nucleotides may comprise other bases such as inosine, deoxyinosine, hypoxanthine may be used.
  • isoteric purine 2′deoxy-furanoside analogs, 2′-deoxynebularine or 2′deoxyxanthosine, or other purine or pyrimidine analogs may also be used.
  • inosine may be used to reduce hybridization specificity
  • diaminopurines may be used to increase hybridization specificity.
  • Adenine and guanine may be modified at positions N3, N7, N9, C2, C4, C5, C6, or C8 and still maintain their hydrogen bonding abilities.
  • Cytosine, thymine and uracil may be modified at positions N1, C2, C4, C5, or C6 and still maintain their hydrogen bonding abilities.
  • Some base analogs have different hydrogen bonding attributes than the naturally occurring bases. For example, 2-amino-2′-dA forms three (3), instead of the usual two (2), hydrogen bonds to thymine (T).
  • Examples of base analogs that have been shown to increase duplex stability include, but are not limited to, 5-fluoro-2′-dU, 5-bromo-2′-dU, 5-methyl-2′-dC, 5-propynyl-2′-dC, 5-propynyl-2′-dU, 2-amino-2′-dA, 7-deazaguanosine, 7-deazadenosine, and N2-Imidazoylpropyl-2′-dG.
  • Nucleotide analogs may be created by modifying and/or replacing a sugar moiety.
  • the sugar moieties of the nucleotides may also be modified by the addition of one or more substituents.
  • one or more of the sugar moieties may contain one or more of the following substituents: amino, alkylamino, araalkyl, heteroalkyl, heterocycloalkyl, aminoalkylamino, O, H, an alkyl, polyalkylamino, substituted silyl, F, Cl, Br, CN, CF 3 , OCF 3 , OCN, O-alkyl, S-alkyl, SOMe, SO 2 Me, ONO 2 , NH-alkyl, OCH 2 CH ⁇ CH 2 , OCH 2 CCH, OCCHO, allyl, O-allyl, NO 2 , N 3 , and NH 2 .
  • the 2′ position of the sugar may be modified to contain one of the following groups: H, OH, OCN, O-alkyl, F, CN, CF 3 , allyl, O-allyl, OCF 3 , S-alkyl, SOMe, SO 2 Me, ONO 2 , NO 2 , N 3 , NH 2 , NH-alkyl, or OCH ⁇ CH 2 , OCCH, wherein the alkyl may be straight, branched, saturated, or unsaturated.
  • the nucleotide may have one or more of its sugars modified and/or replaced so as to be a ribose or hexose (i.e. glucose, galactose) or have one or more anomeric sugars.
  • the nucleotide may also have one or more L-sugars.
  • the sugar may be modified to contain one or more linkers for attachment to other chemicals such as fluorescent labels.
  • the sugar is linked to one or more aminoalkyloxy linkers.
  • the sugar contains one or more alkylamino linkers. Aminoalkyloxy and alkylamino linkers may be attached to biotin, cholic acid, fluorescein, or other chemical moieties through their amino group.
  • Nucleotide analogs or derivatives may have pendant groups attached.
  • Pendant groups serve a variety of purposes which include, but are not limited to, increasing cellular uptake of the molecule, enhancing degradation of the target nucleic acid, and increasing hybridization affinity.
  • Pendant groups can be linked to the binding domains of the catalytic nucleic acid. Examples of pendant groups include, but are not limited to: acridine derivatives (i.e.
  • cross-linkers such as psoralen derivatives, azidophenacyl, proflavin, and azidoproflavin; artificial endonucleases; metal complexes such as EDTA-Fe(II), o-phenanthroline-Cu(I), and porphyrin-Fe(II); alkylating moieties; nucleases such as amino-1-hexanolstaphylococcal nuclease and alkaline phosphatase; terminal transferases; abzymes; cholesteryl moieties; lipophilic carriers; peptide conjugates; long chain alcohols; phosphate esters; amino; mercapto groups; radioactive markers; nonradioactive markers such as dyes; and polylysine or other polyamines.
  • cross-linkers such as psoralen derivatives, azidophenacyl, proflavin, and azidoproflavin
  • artificial endonucleases such as EDTA-Fe(II), o
  • the nucleic acid comprises an oligonucleotide conjugated to a carbohydrate, sulfated carbohydrate, or gylcan.
  • Conjugates may be regarded as a way as to introduce a specificity into otherwise unspecific DNA binding molecules by covalently linking them to a selectively hybridizing oligonucleotide.
  • the binding domains of the catalytic nucleic acid may have one or more of their sugars modified or replaced so as to be ribose, glucose, sucrose, or galactose, or any other sugar. Alternatively, they may have one or more sugars substituted or modified in its 2′ position, i.e. 2′allyl or 2′-O-allyl. An example of a 2′-O-allyl sugar is a 2′-O-methylribonucleotide. Further, the nucleotides of the binding domain may have one or more of their sugars substituted or modified to form an ⁇ -anomeric sugar.
  • a catalytic nucleic acid binding domain may include non-nucleotide substitution.
  • the non-nucleotide substitution includes either abasic nucleotide, polyether, polyamine, polyamide, peptide, carbohydrate, lipid or polyhydrocarbon compounds.
  • abasic or “abasic nucleotide” as used herein encompasses sugar moieties lacking a base or having other chemical groups in place of base at the 1′ position.
  • the nucleotides of the first binding domain comprise at least one modified internucleoside bond.
  • the nucleotides of the second binding domain comprise at least one modified internucleoside bond.
  • the modified internucleoside bond is a phosphorothioate bond.
  • the nucleic acid may comprise modified bonds.
  • the bonds between nucleotides of the catalytic nucleic acid may comprise phosphorothioate linkages.
  • the nucleic acid may comprise nucleotides having moiety may be modified by replacing one or both of the two bridging oxygen atoms of the linkage with analogues such as —NH, —CH 2 , or —S. Other oxygen analogues known in the art may also be used.
  • the phosphorothioate bonds may be stereo regular or stereo random.
  • the PAI-1-encoding mRNA encodes human PAI-1.
  • the human PAI-1-encoding mRNA has the sequence set forth in SEQ ID NO:5.
  • This invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the instant catalytic nucleic acid and a pharmaceutically acceptable carrier.
  • the following pharmaceutically acceptable carriers are set forth, in relation to their most commonly associated delivery systems, by way of example, notwithstanding the fact that the instant pharmaceutical compositions are preferably delivered dermally.
  • Dermal delivery systems include, for example, aqueous and nonaqueous gels, creams, multiple emulsions, microemulsions, liposomes, ointments, aqueous and nonaqueous solutions, lotions, aerosols, hydrocarbon bases and powders, and can contain excipients such as solubilizers, permeation enhancers (e.g., fatty acids, fatty acid esters, fatty alcohols and amino acids), and hydrophilic polymers (e.g., polycarbophil and polyvinylpyrolidone).
  • the pharmaceutically acceptable carrier is a liposome or a transdermal enhancer.
  • liposomes which can be used in this invention include the following: (1) CellFectin, 1:1.5 (M/M) liposome formulation of the cationic lipid N,NI,NII,NIII-tetramethyl-N,NI,NII,NIII-tetrapalmity-spermine and dioleoyl phosphatidylethanolamine (DOPE)(GIBCO BRL); (2) Cytofectin GSV, 2:1 (M/M) liposome formulation of a cationic lipid and DOPE (Glen Research); (3) DOTAP (N-[1-(2,3-dioleoyloxy)-N,N,N-tri-methyl-ammoniumethyl sulfate) (Boehringer Manheim); and (4) Lipofectamine, 3:1 (M/M) liposome formulation of the polycationic lipid DOSPA and the neutral lipid DOPE (GIBCO BRL).
  • DOPE dioleoyl phosphatidylethanolamine
  • Transmucosal delivery systems include patches, tablets, suppositories, pessaries, gels and creams, and can contain excipients such as solubilizers and enhancers (e.g., propylene glycol, bile salts and amino acids), and other vehicles (e.g., polyethylene glycol, fatty acid esters and derivatives, and hydrophilic polymers such as hydroxypropylmethylcellulose and hyaluronic acid).
  • solubilizers and enhancers e.g., propylene glycol, bile salts and amino acids
  • other vehicles e.g., polyethylene glycol, fatty acid esters and derivatives, and hydrophilic polymers such as hydroxypropylmethylcellulose and hyaluronic acid.
  • Injectable drug delivery systems include solutions, suspensions, gels, microspheres and polymeric injectables, and can comprise excipients such as solubility-altering agents (e.g., ethanol, propylene glycol and sucrose) and polymers (e.g., polycaprylactones and PLGA's).
  • Implantable systems include rods and discs, and can contain excipients such as PLGA and polycaprylactone.
  • Oral delivery systems include tablets and capsules. These can contain excipients such as binders (e.g., hydroxypropylmethylcellulose, polyvinyl pyrilodone, other cellulosic materials and starch), diluents (e.g., lactose and other sugars, starch, dicalcium phosphate and cellulosic materials), disintegrating agents (e.g., starch polymers and cellulosic materials) and lubricating agents (e.g., stearates and talc).
  • excipients such as binders (e.g., hydroxypropylmethylcellulose, polyvinyl pyrilodone, other cellulosic materials and starch), diluents (e.g., lactose and other sugars, starch, dicalcium phosphate and cellulosic materials), disintegrating agents (e.g., starch polymers and cellulosic materials) and lubricating agents (e.
  • This invention provides a method of specifically inhibiting the expression of PAI-1 in a cell that would otherwise express PAI-1, comprising contacting the cell with the instant catalytic nucleic acid so as to specifically inhibit the expression of PAI-1 in the cell.
  • This invention provides a method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the instant catalytic nucleic acid effective to specifically inhibit the expression of PAI-1 in the subject's cells.
  • This invention provides a method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the instant pharmaceutical composition effective to specifically inhibit the expression of PAI-1 in the subject's cells.
  • This invention provides a method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the instant pharmaceutical composition effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease.
  • the cardiovascular disease is congestive heart failure. In another embodiment the cardiovascular disease is myocardial infarct. In another embodiment the cardiovascular disease is angina. In another embodiment the cardiovascular disease is myocardial ischemia. In another embodiment the cardiovascular disease is a cardiomyopathy.
  • This invention provides a method of treating a fibrotic disease in a subject involving fibrogenesis which comprises administering to the subject an amount of the instant pharmaceutical composition effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease.
  • the fibrotic disease is a renal disease, a hepatic disease, a disease of the lung, a disease of the skin, or a disease of the eye.
  • the fibrotic disease of the skin is scleroderma or psorasis.
  • an “effective amount” is an amount at least sufficient to treat, reduce, reverse or otherwise inhibit the given disease state.
  • fibrotic diseases this means an amount sufficient to reduce, reverse or otherwise inhibit fibrogenesis.
  • cardiovascular diseases this means an amount sufficient to reduce, reverse or otherwise inhibit cardiomyocyte apoptosis.
  • the effective amount of the instant pharmaceutical compositions can be done based on animal data using routine computational methods.
  • the effective amount contains between about 10 ng and about 100 ⁇ g of the instant nucleic acid molecules per kg body mass. In another embodiment, the effective amount contains between about 100 ng and about 10 ⁇ g of the nucleic acid molecules per kg body mass. In a further embodiment, the effective amount contains between about 1 ⁇ g and about 5 ⁇ g of the nucleic acid molecules per kg body mass. In another embodiment the effective amount contains between about 10 ⁇ g and about 100 ⁇ g of the nucleic acid molecules per kg body mass. In a preferred embodiment the effective amount contains between about 100 ⁇ g and 500 ⁇ g of the nucleic acid molecules per kg body mass. In another embodiment the effective amount contains between about 500 ⁇ g and 1000 ⁇ g of the nucleic acid molecules per kg body mass. In another embodiment the effective amount contains between about 1 mg and 2 mg of the nucleic acid molecules per kg body mass.
  • the compounds of the invention may also be admixed, encapsulated, conjugated or otherwise associated with other molecules, molecule structures or mixtures of compounds, as for example, liposomes, receptor targeted molecules, oral, rectal, topical or other formulations, for assisting in uptake, distribution and/or absorption.
  • Representative United States patents that teach the preparation of such uptake, distribution and/or absorption assisting formulations include, but are not limited to, U.S. Pat. Nos.
  • catalytic nucleic acids it is preferred to administer catalytic nucleic acids to mammals suffering from a cardiovascular disease or a fibrotic disease, in either native form or suspended in a carrier medium in amounts and upon treatment schedules which are effective to therapeutically treat the mammals to reduce the detrimental effects of cardiovascular disease or fibrotic disease.
  • One or more different catalytic nucleic acids or antisense oligonucleotides or analogs thereof targeting different sections of the nucleic acid sequence of PAI-1-encoding mRNA may be administered together in a single dose or in different doses and at different amounts and times depending upon the desired therapy.
  • the catalytic nucleic acids can be administered to mammals in a manner capable of getting the nucleic acids initially into the blood stream and subsequently into cells, or alternatively in a manner so as to directly introduce the catalytic nucleic acids into the cells or groups of cells, for example cardiomyocytes, by such means by electroporation or by direct injection into the heart or by catheter into renal tissue. It is within the scale of a person's skill in the art to determine optimum dosages and treatment schedules for such treatment regimens.
  • the effective amount of catalytic nucleic acid is administered to a human patient in need of inhibition of PAI-1 expression (or inhibition of fibrogenesis) from 1-8 or more times daily or every other day. Dosage is dependent on severity and responsiveness of the effects of the cardiovascular or fibrotic disease to be treated, with a course of treatment lasting from several days to months or until a cure is effected or a reduction of the effects is achieved.
  • the actual effective amount, or dosage, administered may take into account the size and weight of the patient, whether the nature of the treatment is prophylactic, therapeutic in nature, the age, weight, health and sex of the patient, the route of administration, and other factors.
  • compositions may contain suitable excipients and auxiliaries which facilitate processing of the catalytic nucleic acids into preparations which can be used pharmaceutically.
  • the preparations particularly those which can be administered orally and which can be used for the preferred type of administration, such as tablets, dragees, and capsules, and preparations which can be administered rectally, such as suppositories, as well as suitable solutions for administration parenterally or orally, and compositions which can be administered bucally or sublingually, including inclusion compounds, contain from about 0.1 to about 99 percent by weight of active ingredients, together with the excipient.
  • the pharmaceutical preparations of the present invention are manufactured in a manner which is itself well known in the art.
  • the pharmaceutical preparations may be made by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes.
  • the process to be used will depend ultimately on the physical properties of the active ingredient used.
  • Suitable excipients are, in particular, fillers such as sugars, for example, lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example, tricalcium phosphate or calcium hydrogen phosphate as well as binders such as starch, paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone.
  • fillers such as sugars, for example, lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example, tricalcium phosphate or calcium hydrogen phosphate as well as binders such as starch, paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl
  • disintegrating agents may be added, such as the above-mentioned starches as well as carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate.
  • Auxiliaries are flow-regulating agents and lubricants, for example, such as silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol.
  • Dragee cores may be provided with suitable coatings which, if desired, may be resistant to gastric juices.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinylpyrrolidone, polyethylene, glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • suitable cellulose preparations such as acetyl-cellulose phthalate or hydroxypropylmethycellulose phthalate, are used.
  • Dyestuffs and pigments may be added to the tablets of dragee coatings, for example, for identification or in order to characterize different combinations of active compound doses.
  • Other pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol.
  • the push-fit capsules can contain the active compounds in the form of granules which may be mixed with filters such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and optionally, stabilizers.
  • the active compounds are preferably dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of the active compounds with a suppository base.
  • Suitable suppository bases are, for example, natural or synthetic triglycerides, paraffin hydrocarbons, polyethylene glycols or higher alkanols.
  • gelatin rectal capsules which consist of a combination of the active compounds with a base.
  • Possible base materials include, for example liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
  • Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble or water-dispersible form.
  • suspensions of the active compounds as appropriate oily injection suspensions may be administered.
  • Suitable lipophilic solvents or vehicles include fatty oils, for example, ethyl oleate or triglycerides.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran.
  • the suspension may also contain stabilizers.
  • catalytic nucleic acids of the present invention may also be administered encapsulated in liposomes or immunoliposomes, which are pharmaceutical compositions wherein the active ingredient is contained either dispersed or variously present in corpuscles consisting of aqueous concentric layers adherent to lipidic layers.
  • liposomes are especially active in targeting the oligonucleotides to liver cells.
  • the active ingredient depending upon its solubility, may be present both in the aqueous layer and in the lipidic layer, or in what is generally termed a liposomic suspension.
  • the hydrophobic layer generally but not exclusively, comprises phospholipids such as lecithin and sphingomyelin, steroids such as cholesterol, more or less ionic surfactants such as dicetylphosphate, stearylamine, or phosphatidic acid, and/or other materials of a hydrophobic nature.
  • the diameters of the liposomes generally range from about 15 nm to about 5 microns.
  • This invention also provides an oligonucleotide comprising consecutive nucleotides that hybridizes with a PAI-1-encoding mRNA under conditions of high stringency and is between 8 and 40 nucleotides in length. In different embodiments the oligonucleotide is between 40 and 80 nucleotides in length.
  • the hybridization of antisense oligonucleotides with PAI-1-encoding mRNA interferes with one or more of the normal functions of PAI-1-encoding mRNA.
  • the functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA.
  • a human PAI-1 antisense oligonucleotide specifically hybridizes under given stringent conditions with targets on the human PAI-1 mRNA molecule and in doing so inhibits the translation thereof into PAI-1 protein.
  • the functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA.
  • a PAI-1 antisense oligonucleotide specifically hybridizes under given stringent conditions with targets on the PAI-1-encoding mRNA molecule and in doing so inhibits the translation thereof into PAI-1.
  • messenger RNA includes not only the information to encode a protein using the three letter genetic code, but also associated ribonucleotides which form a region known to such persons as the 5′-untranslated region, the 3′-untranslated region, the 5′ cap region and intron/exon junction ribonucleotides.
  • catalytic nucleic acids or antisense oligonucleotides may be formulated in accordance with this invention which are targeted wholly or in part to these associated ribonucleotides as well as to the informational ribonucleotides.
  • the antisense oligonucleotides may therefore be specifically hybridizable with a transcription initiation site region, a translation initiation codon region, a 5′ cap region, an intron/exon junction, coding sequences, a translation termination codon region or sequences in the 5′- or 3′-untranslated region.
  • the catalytic nucleic acids may specifically cleave a transcription initiation site region, a translation initiation codon region, a 5′ cap region, an intron/exon junction, coding sequences, a translation termination codon region or sequences in the 5′- or 3′-untranslated region.
  • the translation initiation codon is typically 5′-AUG (in transcribed mRNA molecules; 5′-ATG in the corresponding DNA molecule).
  • 5′-AUG in transcribed mRNA molecules
  • 5′-ATG in the corresponding DNA molecule
  • a minority of genes have a translation initiation codon having the RNA sequence 5′-GUG, 5′UUG or 5′-CUG, and 5′-AUA, 5′-ACG and 5′-CUG have been shown to function in vivo.
  • the term “translation initiation codon” can encompass many codon sequences, even though the initiator amino acid in each instance is typically methionine in eukaryotes.
  • translation initiation codon refers to the codon or codons that are used in vivo to initiate translation of an mRNA molecule transcribed from a gene encoding PAI-1, regardless of the sequence(s) of such codons.
  • a translation termination codon of a gene may have one of three sequences, i.e., 5′-UAA, 5′-UAG and 5′-UGA (the corresponding DNA sequences are 5′-TAA, 5′-TAG and 5′-TGA, respectively).
  • the term “translation initiation codon region” refers to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′) from a translation initiation codon. This region is one preferred target region.
  • translation termination codon region refers to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′) from a translation termination codon. This region is also one preferred target region.
  • Other preferred target regions include the 5′ untranslated region (5′UTR), known in the art to refer to the portion of an mRNA in the 5′ direction from the translation initiation codon, and thus including nucleotides between the 5′ cap site and the translation initiation codon of an mRNA or corresponding nucleotides on the gene, and the 3′ untranslated region (3′UTR), known in the art to refer to the portion of an mRNA in the 3′ direction from the translation termination codon, and thus including nucleotides between the translation termination codon and 3′ end of an mRNA or corresponding nucleotides on the gene.
  • 5′UTR 5′ untranslated region
  • 3′UTR 3′ untranslated region
  • mRNA splice sites may also be preferred target regions, and are particularly useful in situations where aberrant splicing is implicated in disease, or where an overproduction of a particular mRNA splice product is implicated in disease. Aberrant fusion junctions due to rearrangements or deletions may also be preferred targets.
  • antisense oligonucleotides can be chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired disruption of the function of the molecule.
  • “Hybridization”, in the context of this invention means hydrogen bonding, also known as Watson-Crick base pairing, between complementary bases, usually on opposite nucleic acid strands or two regions of a nucleic acid strand. Guanine and cytosine are examples of complementary bases which are known to form three hydrogen bonds between them. Adenine and thymine are examples of complementary bases which form two hydrogen bonds between them.
  • Specifically hybridizable and “complementary” are terms which are used to indicate a sufficient degree of complementarity such that stable and specific binding occurs between the DNA or RNA target and the antisense oligonucleotide.
  • catalytic nucleic acids are synthesized once cleavage target sites on the PAI-1 mRNA molecule have been identified, e.g. any purine:pyrimidine consensus sequences in the case of DNA enzymes.
  • Catalytic nucleic acids and antisense oligonucleotides targeted to disrupting polymorphisms and mutants of the PAI-1 maybe similarly made as described above based on the polymorphic or mutant PAI-1 mRNA sequence.
  • At least one internucleoside linkage within the instant oligonucleotide comprises a phosphorothioate linkage.
  • Antisense oligonucleotide molecules synthesized with a phosphorothioate backbone have proven particularly resistant to exonuclease damage compared to standard deoxyribonucleic acids, and so they are used in preference.
  • a phosphorothioate antisense oligonucleotide for PAI-1 mRNA can be synthesized on an Applied Biosystems (Foster City, Calif.) model 380B DNA synthesizer by standard methods.
  • oligodeoxynucleotides can be base deblocked in ammonium hydroxide at 60° C. for 8 h and purified by reversed-phase HPLC [0.1M triethylammonium bicarbonate/acetonitrile; PRP-1 support]. Oligomers can be detritylated in 3% acetic acid and precipitated with 2% lithiumperchlorate/acetone, dissolved in sterile water and reprecipitated as the sodium salt from 1 M NaCl/ethanol. Concentrations of the full length species can be determined by UV spectroscopy. Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and alkylated derivatives.
  • Preferred modified oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3T-alkylene phosphonates, 5′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and borano-phosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3′ to 3′, 5′ to 5′ or 2′ to 2′ linkage.
  • Preferred oligonucleotides having inverted polarity comprise a single 3′ to 3′ linkage at the 3′-most internucleotide linkage i.e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof).
  • Various salts, mixed salts and free acid forms are also included.
  • Representative United States patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos.
  • nucleotides of the instant oligonucleoptide comprise at least one deoxyribonucleotide. In another embodiment the nucleotides comprise at least one ribonucleotide. Such deoxyribonucleotides or ribonucleotides can be modified or derivatized as described hereinabove.
  • the PAI-1-encoding mRNA encodes human PAI-1.
  • the human PAI-1-encoding mRNA comprises consecutive nucleotides, the sequence of which is set forth in SEQ ID NO:5.
  • This invention further provides a method of treating a subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit expression of a PAI-1 in the subject so as to thereby treat the subject.
  • This invention further provides a method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease.
  • This invention further provides a method of treating a fibrotic disease in a subject involving fibrogenesis in the subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease.
  • Antisense oligonucleotides can be administered by intravenous injection, intravenous drip, subcutaneous, intraperitoneal or intramuscular injection, orally or rectally. Human pharmacokinetics of certain antisense oligonucleotides have been studied. See (37) incorporated by reference in its entirety.
  • This invention provides all of the instant methods wherein the subject is a mammal.
  • the mammal is a human being.
  • E1 SEQ ID NO:2
  • E2 SEQ ID NO:4
  • E3 SEQ ID NO:3
  • SEQ ID NO:1 15-nucleotide catalytic domains
  • E0 SEQ ID NO:7
  • the nucleotide sequence in the two flanking arms of E2 was scrambled without altering the catalytic domain (FIG. 1 a ).
  • the 3′ terminus of each molecule was capped with an inverted 3′-3′-linked thymidine for resistance to 3′-to-5′ exonuclease digestion.
  • the 21-base oligonucleotide S1 (SEQ ID NO:8), synthesized from human PAI-1 mRNA and labeled at the 5′ end with 32P, was cleaved within 2 minutes when cultured with E1 at 10:1 substrate:enzyme excess, with maximal cleavage occurring by 2 hours.
  • the 10-nucleotide cleavage product is consistent with the size of the 32P-labeled fragment at the 5′ end.
  • E1 also cleaved larger 32P-labeled fragments of human PAI-1 mRNA, prepared by in vitro transcription, in a time- and concentration-dependent manner, FIG. 1 c .
  • a 520 nucleotide transcript was maximally cleaved by 2-4 hours to expected cleavage products of 320 and 200 nucleotides.
  • a similar dose-dependency was seen with E3, with maximal cleavage of human PAI-1 mRNA transcripts occurring at the highest concentration used, 20 ⁇ M.
  • the sequence-specific nature of the DNA enzymatic cleavage is shown in FIG. 1 d , where the control DNA enzyme E0, containing an identical catalytic domain to E1 and E3, but scrambled sequences in the flanking arms, caused no cleavage of human PAI-1 mRNA transcripts. Pre-heating the transcript to 72° C. for 10 minutes prior to incubation with E1, but not E0, further increased cleavage.
  • FIG. 2 a The DNA enzyme E2 cleaved the 23-base oligonucleotide S2, synthesized from the sequence of rat PAI-1 mRNA, in a dose- and time-dependent manner, FIG. 2 a .
  • E2 also cleaved a rat PAI-1 mRNA transcript in a dose-dependent manner by 2-4 hours to give the 156 nucleotide cleavage product, FIG. 2 b .
  • neither the scrambled control DNA enzyme E0 nor E3 cleaved the rat PAI-1 mRNA transcript.
  • 3 b shows the effect of endothelial cell transfection with E2 DNA enzyme on TGF-beta mediated induction of PAI-1 protein.
  • Endothelial cells transfected with scrambled DNA enzyme demonstrated approximately 50% increase in cytoplasmic PAI-1 protein as detected by Western blot. In contrast, this effect was almost completely is abrogated by transfection with the PAI-1 DNA enzyme E2.
  • DNA enzymes with a thymidine in the correct orientation at the 3′ end are significantly degraded by factors in serum (1-5% concentration) within 24 hours (14), we investigated the protective effect of an inverted thymidine at the 3′ end on serum-dependent nucleolytic degradation of PAI-1 DNA enzymes.
  • the DNA enzyme E1 was labeled with 32P and incubated for 3-24 hours with human umbilical vein endothelial cell (HUVEC) monolayers cultured in medium containing physiologic (2%) or supraphysiologic (20%) serum concentrations As shown in FIG.
  • HUVEC human umbilical vein endothelial cell
  • PAI-1 expression in infarcted rat hearts was dramatically altered, with 71% reduction in PAI-1 expression by macrophages at the peri-infarct region (p ⁇ 0.01), FIG. 4 a .
  • Inhibition of PAI-1 expression at this site was highly specific since PAI-1 expression within macrophages in the center of the infarct zone remained unaffected after E2 injection, and injection with the scrambled DNA enzyme E0 did not reduce peri-infarct PAI-1 expression.
  • E2 Protects Cardiomyocytes at the Peri-Infarct Region Against Apoptosis.
  • the renin-angiotensin system (RAS) in progressive renal disease has been extensively investigated, indicating multiple actions beyond hemodynamic and salt/water homeostasis.
  • the RAS is now recognized to be linked to induction of plasminogen activator inhibitor-1 (PAI-1) likely via both the type 1 (AT1) and type 4 (AT4) receptors, thus, promoting both thrombosis and fibrosis (39).
  • Plasminogen activator inhibitor type 1 is thus a suitable target in renal fibrogenesis.
  • the progression of renal lesions to fibrosis involves several mechanisms, among which the inhibition of extracellular matrix (ECM) degradation appears to play an important role.
  • ECM extracellular matrix
  • PAI-1 as the main inhibitor of plasminogen activation, regulates fibrinolysis and the plasmin-mediated matrix metalloproteinase activation. PAI-1 is also a component of the ECM, where it binds to vitronectin. PAI-1 is not expressed in the normal human kidney but is strongly induced in various forms of kidney diseases, leading to renal fibrosis and terminal renal failure. Thrombin, angiotensin II, and transforming growth factor-beta are potent in vitro and in vivo agonists in increasing PAI-1 synthesis.
  • Several experimental and clinical studies support a role for PAI-1 in the renal fibrogenic process occurring in chronic glomerulonephritis, diabetic nephropathy, focal segmental glomerulosclerosis, and other fibrotic renal diseases (40).
  • End-stage renal disease comprises an enormous public health burden, with an incidence and prevalence that are increasingly on the rise. This escalating prevalence suggests that newer therapeutic interventions and strategies are needed to complement current therapeutic approaches. Although much evidence demonstrates conclusively that angiotensin II mediates progressive renal disease, recent evidence also implicates aldosterone as an important pathogenetic factor in progressive renal disease.
  • Aldosterone may promote fibrosis by several mechanisms, including plasminogen activator inhibitor-1 (PAI-1) expression and consequent alterations of vascular ribrinolysis, by stimulation of transforming growth factor-betal (TGF-betal), and by stimulation of reactive oxygen species (ROS) (41). Therefore it is expected that PAI-1 is a suitable target for therapeutic intervention into renal fibrosis.
  • PAI-1 plasminogen activator inhibitor-1
  • TGF-betal transforming growth factor-betal
  • ROS reactive oxygen species
  • PAI-1 plasminogen activator inhibitor-1
  • TGF-beta transforming growth factor beta
  • HSCs myofibroblast-like cells derived from hepatic stellate cells
  • PAI-1 plasminogen activator inhibitor type 1
  • COL1A2 alpha2(I) procollagen
  • Increased PAI-1 together with uPA, uPAR and tPA are associated with overall inhibition of matrix degradation in cirrhotic liver.
  • Hepatic stellate cells are an important source of PAI-1 during liver fibrosis.
  • PAI-1 levels in bronchoalveolar lavage (BAL) supernatant fluids are significantly higher in idiopathic pulmonary fibrosis (IPF) patients than in normal subjects. Increased procoagulant and antifibrinolytic activities in the lungs with idiopathic pulmonary fibrosis. Therefore it is expected that PAI-1 is a suitable target for therapeutic intervention into fibrosis of the lung.
  • BAL bronchoalveolar lavage
  • IPF idiopathic pulmonary fibrosis
  • cataracts of the eye may be treated by reversing the symptomatic fibrosis that occurs, thought to be due to ischemia, by the mechanism described hereinabove.
  • DNA enzymes and RNA substrates DNA enzymes with 3′-3′ inverted thymidine were synthesized by Integrated DNA technologies (Coralville, Iowa) and purified by RNase-free IE-HPLC or RP-HPLC. The short RNA substrates corresponding to target DNA enzyme sequences were chemically synthesized followed by RNAse-free PAGE purification and also made by in vitro transcription from a DNA template.
  • a 32P-labeled-nucleotide human PAI-1 RNA transcript was prepared by in vitro transcription (SP6 polymerase, Promega). 20 ml transcription reaction consisted of 4 ⁇ l 5 ⁇ buffer, 2 ⁇ l DTT, 1 ⁇ l RNasin inhibitor, 4 ⁇ l NTP mixture (1 ⁇ l A,G,C, and 1 ul H 2 O), 100 ⁇ M UTP, 2 ⁇ l template (0.3 ⁇ g/ ⁇ l), a-32P-UTP (10 ⁇ ci/ ⁇ l) and 1 ⁇ l SP6 polymerase (20 u/ ⁇ l). Reaction time was 1 hour at 32° C. Unincorporated label and short nucleotides ( ⁇ 350base) were seperated from radiolabeled species by centrigugation on Chromaspin-200 columns (Clontech, Palo Alto, Calif.).
  • Cleavage reactions synthetic RNA substrate was end-labeled with 32P using T4 polynucleotide kinase.
  • Cleavage reaction system included 60 mM Tris-HCl (pH 7.5), 10 mM MgCl 2 , 150 mM NaCl, 0.5 ⁇ M 32p-labled RNA oligo., 0.05-5 ⁇ M DNA enzyme.
  • reaction system contained 1% of PAI-1 transcripts, 25 mM Tris-HCl (pH 7.5), 5 mM MgCl 2 , 100 mM NaCl and 0.2-20 ⁇ M DNA enzyme. Reactions were allowed to proceed at 37° C.
  • DNA enzyme stability in serum DAN enzymes were radiolabled using T4 polynucleotide kinase.
  • Labeling reaction (20 ⁇ l of volume) consists of 1 ⁇ l 20 ⁇ M DNA enzyme, 2 ⁇ l 10 ⁇ kinase buffer, 1 ⁇ l T4 PNK (10 u/ ⁇ l), 10 ⁇ l H 2 O, and 6 ⁇ l 32P-ATP (3000 uCi/mmol, 10 uCi/ ⁇ l). Reaction time is 30 minutes at 37° C.
  • HUVEC were cultured in media (Clonetic) containing 2% or 20% FCS, and radiolabeled oligomers were added at a final concentration of 100 nM.
  • HUVEC endothelial cells were obtained from Clonetic (USA) and grown in medium containing 2%FCS, 100 ug/ml streptomycin and 100 IU/ml penicillin at 37° C. in a humidified atmosphere of 5% CO2. HUVEC were used in experiments between passage 6 and 8. For transfection of DNA enzyme, rat endothelial cells (EC) were harvested and seeded into each well of 6-well plates ( ⁇ 1 ⁇ 10 5 cells/well).
  • Subconfluent (70-80%) EC were washed twice with 1 ml HEPES buffer (pH 7.4) and transfected using 0.5 ml of serum-free medium containing 1 ⁇ M test molecule (E2 or E0) and 20 ⁇ g/ml cationic lipids (DOTAP). 3 hours after incubation, 0.5 ml of 2% serum medium were added to each well; 3 hours later, TGFb was added to half the wells at a final concentration of 1.8 ng/ml. Transfected cells continued to be incubated for 8 hrs and were lysed to isolate RNA (for RT-PCR) and Protein (for Western blot) using Trizol reagent (Life Technologies).
  • RT-PCR Total RNA from each well of 6-well plates were isolated using 1 ml Trizol reagent and dissolved in 20 ⁇ l Depc-treated H 2 O. 2 ⁇ l samples were used to perform reverse transcription in 20 ⁇ l reaction system, and 4 ⁇ l products were then used as templates to amplify PAI-1 or human GAPDH in 50 ⁇ l PCR system (5 ⁇ l 10 ⁇ buffer, 1 ⁇ l dNTP, 0.25 ⁇ l Taq polymerase, 1 ⁇ l forward and reverse primer, 38 ⁇ l H 2 O, 2 ⁇ l 32pdCTP (3000 ci/mmol, 10 uci/ul).
  • the reaction conditions were 95° C.-2 min (predenatured), 95° C.-0.5 min, 58° C.-1 min, 72° C.-2 min, 35 cycles, 72° C.-10 min.
  • Human GAPDH was used as internal control (forward Primer 5′TGAAGGTCGGAGTCAACGGATTTG3′; SEQ ID NO:13, reverse primer 5′CATGTGGGCCATGAGGTCCACCAC3′; SEQ ID NO:14), its PCR products being 452 base.
  • proteins were transferred to nitrocellulose membrane (Protran, Schleicler & Schuell) by electrotransfer and blocked for at least one hour at room temperature with 5% (W/V) BSA in TBS-T buffer (0.1M Tris-base (pH 7.5), 0.15M Nacl and 0.1% Tween-20) Following this step, membranes were immunoblotted with goat IgG polyclonal anti-PAI antibodies (Santa Cruz biotech) and then visualised by the ECL-system (Amershan) using horseradish peroxidase conjugated anti-goat IgG (Sigma).
  • tissue sections were then digested with Proteinase K (10 ⁇ g/ml in Tris/HCL) for 30 minutes at 37° C.
  • the slides were then washed 3 times in PBS and incubated with 50 ⁇ l of the TUNEL reaction mixture (TdT and fluorescein-labeled dUTP) and incubated in a humid atmosphere for 60 minutes at 37° C.
  • TdT was eliminated from the reaction mixture.
  • the sections were then incubated for 30 minutes with an antibody specific for fluorescein-conjugated alkaline phosphatase (AP) (Boehringer Mannheim, Mannheim, Germany).
  • AP fluorescein-conjugated alkaline phosphatase
  • the TUNEL stain was visualized with a substrate system in which nuclei with DNA fragmentation stained blue, (BCIP/NBT substrate system, DAKO, Carpinteria, Calif.). The reaction was terminated following three minutes of exposure with PBS. To determine the proportion of blue-staining apoptotic nuclei within myocytes, tissue was counterstained with a monoclonal antibody specific for desmin. Endogenous peroxidase was blocked by using a 3% hydrogen perioxidase solution in PBS for 15 minutes, followed by washing with 20% goat serum solution. An anti-desmin antibody (Sigma, Saint Louis, Mo.) was incubated overnight (1:100) at 40° C.
  • BCIP/NBT substrate system DAKO, Carpinteria, Calif.
  • the percentage of apoptotic myocytes was determined by means of an apoptotic index; the apoptotic index was calculated by dividing the number of positive staining myocyte nuclei by the total number of myocyte nuclei and multiplying by 100. Stained cells at the edges of the tissue were not counted. An apoptotic index of 1 or less was considered to indicate the absence of apoptosis.
  • RNAdraw an integrated program for RNA secondary structure calculation and analysis under 32-bit Microsoft Windows. Comput Appl Biosci 12:247-9.

Abstract

The present invention provides DNAzymes and ribozymes that specifically cleave PAI-1-encoding mRNA. The present invention also provides antisense oligonucleotides that specifically inhibit translation of PAI-1-encoding mRNA. The invention also provides various methods of inhibiting the expression of PAI-1. Finally the invention provides pharmaceutical compositions containing the instant DNAzymes, ribozymes and antisense oligonucleotides as active ingredients.

Description

  • Throughout this application, various publications are referenced in parentheses by arabic numbers. Full citations for these references may be found at the end of the specification immediately preceding the claims. The disclosures of these publications in their entireties are hereby incorporated by reference into this application to more fully describe the state of the art to which this invention pertains. [0001]
  • BACKGROUND
  • After a myocardial infarction, the subsequent cardiomyocyte necrosis stimulates a wound healing response characterized by migration of inflammatory cells into the affected myocardium, extracellular matrix degradation, and neovascularization. Each of these components appears to require activation of latent metalloproteinases by plasmin, which is derived from plasminogen through activation by urokinase-type plasminogen activator (u-PA) expressed on the surface of the infiltrating bone-marrow derived cells (1-3). More recently, it has been suggested that the role of cell surface u-PA may be to facilitate exposure of cryptic cell attachment sites necessary for cell migration (4). This appears to involve direct interaction between u-PA and plasminogen activator inhibitor-1 (PAI-1), which, in its native state, is complexed to vitronectin (4,5). Reaction of PAI-1 with a proteinase, such as u-PA, results in a rapid conformational change that causes it to dissociate from vitronectin and increase its affinity for the low density lipoprotein receptor (6), leading to its clearance and degradation. Removal of PAI-1 from vitronectin exposes the epitope on vitronectin necessary for binding to another of its ligands, the integrin alpha v beta 3 (4,7). Since interactions between the cell surface integrin [0002] alpha v beta 3 and tissue vitronectin have been shown to be important in the development of angiogenesis (4,8,9), we hypothesized that excessive PAI-1 protein expression after myocardial infarction might prevent optimal neovascularization by bone marrow-derived angioblasts and that inhibition of PAI-1 expression would promote neovascularization.
  • In order to develop an approach to inhibit PAI-1 expression which would have clinical applicability, we examined various potential strategies for inhibiting specific mRNA activity. [0003]
  • Antisense oligonucleotides hybridize with their complementary target site in mRNA, blocking translation to protein by sterically inhibiting ribosome movement or by triggering cleavage by endogenous RNAse H (10). Although current constructs are made more resistant to degradation by serum through phosphorothioate linkages, non-specific biological effects due to “irrelevant cleavage” of non-targeted mRNA remains a major concern (11). [0004]
  • Ribozymes are naturally-occurring RNA molecules that contain catalytic sites, making them more potent agents than antisense oligonucleotides. However, wider use of ribozymes has been hampered by their susceptibility to chemical and enzymatic degradation and restricted target site specificity (12). [0005]
  • A new generation of catalytic nucleic acids has been described containing DNA molecules with catalytic activity for specific RNA sequences (13-16). These DNA enzymes exhibit greater catalytic efficiency than hammerhead ribozymes, producing a rate enhancement of approximately 10 million-fold over the spontaneous rate of RNA cleavage, offer greater substrate specificity, are more resistant to chemical and enzymatic degradation, and are far cheaper to synthesize. [0006]
  • SUMMARY
  • This invention provides a catalytic nucleic acid that specifically cleaves an mRNA encoding a Plasminogen Activator Inhibitor-1 (PAI-1) comprising, in 5′ to 3′ order: [0007]
  • (a) consecutive nucleotides defining a first binding domain of at least 4 nucleotides; [0008]
  • (b) consecutive nucleotides defining a catalytic domain located contiguous with the 3′ end of the first binding domain, and capable of cleaving the PAI-1-encoding mRNA at a predetermined phosphodiester bond; and [0009]
  • (c) consecutive nucleotides defining a second binding domain of at least 4 nucleotides located contiguous with the 3′ end of the catalytic domain, [0010]
  • wherein the sequence of the nucleotides in each binding domain is complementary to a sequence of ribonucleotides in the PAI-1-encoding mRNA and wherein the catalytic nucleic acid hybridizes to and specifically cleaves the PAI-1-encoding mRNA. [0011]
  • This invention further provides a pharmaceutical composition comprising the instant catalytic nucleic acid and a pharmaceutically acceptable carrier. [0012]
  • This invention further provides method of specifically inhibiting the expression of PAI-1 in a cell that would otherwise express PAI-1, comprising contacting the cell with the instant catalytic nucleic acid so as to specifically inhibit the expression of PAI-1 in the cell. [0013]
  • This invention further provides a method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the instant catalytic nucleic acid effective to specifically inhibit the expression of PAI-1 in the subject's cells. [0014]
  • This invention further provides a method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the instant pharmaceutical composition effective to specifically inhibit the expression of PAI-1 in the subject's cells. [0015]
  • This invention further provides a method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the instant pharmaceutical composition effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease. [0016]
  • This invention further provides a method of treating a fibrotic disease in a subject involving fibrogenesis which comprises administering to the subject an amount of the instant pharmaceutical composition effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease. [0017]
  • This invention further provides an oligonucleotide comprising consecutive nucleotides that hybridizes with a PAI-1-encoding mRNA under conditions of high stringency and is between 8 and 40 nucleotides in length. This invention further provides the instant oligonucleotide, wherein the human PAI-1-encoding mRNA comprises consecutive nucleotides, the sequence of which is set forth in SEQ ID NO:5. [0018]
  • This invention further provides a method of treating a subject which comprises administering to the subject an amount of the instant oligonucleotide of effective to inhibit expression of a PAI-1 in the subject so as to thereby treat the subject. [0019]
  • This invention further provides a method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease. [0020]
  • This invention further provides a method of treating a fibrotic disease in a subject involving fibrogenesis in the subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease. [0021]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. [0022] 1(A)-(D): (A) This figure shows three DNA enzymes, termed E1 (SEQ ID NO:2), E2 (SEQ ID NO:4), and E3 (SEQ ID NO:3), containing identical 15-nucleotide catalytic domains (SEQ ID NO:1) flanked by two arms of eight nucleotides (E1, E2) or nine nucleotides (E3) with complementarity to human (E1, E3) or rat (E2) PAI-1 mRNA. This figure also shows the control DNA enzyme E0 (SEQ ID NO:7), an oligonucleotide S1 (SEQ ID NO:8), an oligonucleotide transcript (SEQ ID NO:9) and oligonucleotide S2 (SEQ ID NO:10). (B) This figure shows the 21-base oligonucleotide S1 (SEQ ID NO:8), synthesized from human PAI-1 mRNA and labeled at the 5′ end with 32P, was cleaved within 2 minutes when cultured with E1 at 10:1 substrate:enzyme excess, with maximal cleavage occurring by 2 hours. (C) This figure shows E1 also cleaved larger 32P-labeled fragments of human PAI-1 mRNA, prepared by in vitro transcription, in a time- and concentration-dependent manner. (D) This figure shows the sequence-specific nature of the DNA enzymatic cleavage: the control DNA enzyme E0, containing an identical catalytic domain to E1 and E3, but scrambled sequences in the flanking arms, caused no cleavage of human PAI-1 mRNA transcripts.
  • FIGS. [0023] 2(A)-(D): (A) This figure shows that the DNA enzyme E2 cleaved the 23-base oligonucleotide S2 (SEQ ID NO:10), synthesized from the sequence of rat PAI-1 mRNA, in a dose- and time-dependent manner. (B) This figure shows that E2 also cleaved a rat PAI-1 mRNA transcript in a dose-dependent manner by 2-4 hours to give the 156 nucleotide cleavage product. (C) This figure shows the rat S2 PAI-1 mRNA transcript (SEQ ID NO:10) differs by only one nucleotide from the human mRNA PAI-1 transcript (SEQ ID NO:9) which can be cleaved by E3.
  • FIGS. [0024] 3(A)-(D): (A) This figure shows Densitometric analysis of RT-PCR products following reverse transcription of cellular mRNA: E2 inhibited TGF-beta inducible steady-state mRNA levels in cultured rat endothelium by 52% relative to the E0 scrambled DNA.
  • (B) This figure shows the effect of endothelial cell transfection with E2 DNA enzyme on TGF-beta mediated induction of PAI-1 protein. Endothelial cells transfected with scrambled DNA enzyme demonstrated approximately 50% increase in cytoplasmic PAI-1 E0 protein as detected by Western blot. In contrast, this effect was almost completely abrogated by transfection with the PAI-1 DNA enzyme E2. (C) This figure shows that whereas the DNA enzyme was almost completely degraded within 6 hours of cell culture in medium containing 20% serum, it remained intact during the entire 24 hours of cell culture with medium containing 2% serum. (D) This figure shows that HUVEC transfected with E1 or E3 DNA enzymes and cultured for 12 hours in 2% serum demonstrated 20% and 28% reduction in TGF-beta dependent PAI-1 activity compared with transfection with the scrambled DNA enzyme E0 (both p<0.01). [0025]
  • FIGS. [0026] 4(A)-(C): (A) This figure shows that at two weeks after injection with E2, PAI-1 expression in infarcted rat hearts was dramatically altered, with 71% reduction in PAI-1 expression by macrophages at the peri-infarct region (p<0.01). (B) and (C) show injection of E2 and angioblasts increased the number of CD31 positive capillaries.
  • FIGS. [0027] 5A-C: (A) This figure shows inhibition of cardiomyocyte apoptosis in the peri-infarct region by E2. (B) This figure shows the percentage recovery in left ventricular ejection fraction (LVEF) after treatment with E2 and after treatment with E2 and angioblasts.
  • FIG. 6: This figure shows the DNA sequence encoding human Plasminogen Activator Inhibitor-1 protein (SEQ ID NO:5). [0028]
  • FIG. 7: This figure shows the DNA sequence encoding rat Plasminogen Activator Inhibitor-1 protein (SEQ ID NO:15). [0029]
  • FIG. 8: This figure shows the amino acid sequence of human Plasminogen Activator Inhibitor-1 protein (SEQ ID NO:6). [0030]
  • FIG. 9: This figure shows the amino acid sequence of rat Plasminogen Activator Inhibitor-1 protein (SEQ ID NO:16). [0031]
  • FIG. 10: This figure shows possible 5′-AT-3′ cleavage sites on human PAI-1 mRNA (shown as corresponding DNA, SEQ ID NO:5) for catalytic deoxyribonucleic acids. Bold indicates the protein coding region, capital letters indicate consensus cleavage sites. [0032]
  • FIG. 11: This figure shows possible 5′-AC-3′ cleavage sites on human PAI-1 mRNA (shown as corresponding DNA, SEQ ID NO:5) for catalytic deoxyribonucleic acids. Bold indicates the protein coding region, capital letters indicate consensus cleavage sites. [0033]
  • FIG. 12: This figure shows possible cleavage sites on human PAI-1 mRNA coding region (shown as corresponding DNA, SEQ ID NO:17) for catalytic hammerhead ribonucleic acids. Uppercase “T” represents cleavage site. [0034]
  • DETAILED DESCRIPTION
  • As used herein, and unless stated otherwise, each of the following terms shall have the definition set forth below. [0035]
  • “Administering” shall mean any of the various methods and delivery systems known to those skilled in the art. The administering can be performed, for example, via implant, transmucosally, transdermally and subcutaneously, orally, parenterally, topically, by cardiac injection, by inhalation, by catheter e.g. retrograde ureteric catheter, by intra-arterial injection. [0036]
  • “Catalytic” shall mean the functioning of an agent as a catalyst, i.e. an agent that increases the rate of a chemical reaction without itself undergoing a permanent structural change. [0037]
  • “Consensus sequence” shall mean a nucleotide sequence of at least two residues in length between which catalytic nucleic acid cleavage occurs. For example, consensus sequences include for catalytic deoxyribonucleic acids are purine:pyrimidine e.g. “A:U” and “G:U”. [0038]
  • “Plasminogen Activator Inhibitor-1 Protein” shall mean the protein encoded by the nucleotide sequence identified as (SEQ ID NO:5) and having the amino acid sequence shown in SEQ ID NO:6, when identified as human and the protein encoded by the nucleotide sequence identified as SEQ ID NO:15 and having the amino acid sequence shown in SEQ ID NO:16 when identified as originating from rat, and any variants of either thereof, whether artificial or naturally occurring. Variants include, without limitation, homologues, post-translational modifications, mutants and polymorphisms. [0039]
  • “Plasminogen Activator Inhibitor-1 mRNA” shall mean a mRNA molecule comprising a sequence which encodes Plasminogen Activator Inhibitor-1 Protein. Plasminogen Activator Inhibitor-1 mRNA includes, without limitation, protein-encoding sequences as well as the 5′ and 3′ non-protein-encoding sequences. [0040]
  • “Hybridize” shall mean the annealing of one single-stranded nucleic acid molecule to another nucleic acid molecule based on sequence complementarity. The propensity for hybridization between nucleic acids depends on the temperature and ionic strength of their milieu, the length of the nucleic acids and the degree of complementarity. The effect of these parameters on hybridization is well known in the art (see 38). [0041]
  • “Inhibit” shall mean to slow, stop or otherwise impede. [0042]
  • “Stringent conditions” or “Stringency”, shall refer to the conditions for hybridization as defined by the nucleic acid, salt, and temperature. These conditions are well known in the art and may be altered. Numerous equivalent conditions comprising either low or high stringency depend on factors such as the length and nature of the sequence (DNA, RNA, base composition), nature of the target (DNA, RNA, base composition), milieu (in solution or immobilized on a solid substrate), concentration of salts and other components (e.g., formamide, dextran sulfate and/or polyethylene glycol), and temperature of the reactions (within a range from about 5° C. below the melting temperature of the probe to about 20° C. to 25° C. below the melting temperature). One or more factors be may be varied to generate conditions of either low or high stringency different from, but equivalent to, the above listed conditions. As will be understood by those of skill in the art, the stringency of hybridization may be altered in order to identify or detect identical or related polynucleotide sequences. [0043]
  • “Nucleic acid” shall include any nucleic acid, including, without limitation, DNA, RNA, oligonucleotides, or polynucleotides, and analogs or derivatives thereof. The nucleotides that form the nucleic acid may be nucleotide analogs or derivatives thereof. The nucleic acid may incorporate non nucleotides. [0044]
  • “Nucleotides” shall include without limitation nucleotides and analogs or derivatives thereof. For example, nucleotides may comprise the bases A, C, G, T and U, as well as derivatives thereof. Derivatives of these bases are well known in the art, and are exemplified in PCR Systems, Reagents and Consumables (Perkin Elmer Catalogue 1996-1997, Roche Molecular Systems, Inc., Branchburg, N.J., USA). [0045]
  • “Pharmaceutically acceptable carrier” shall mean any of the various carriers known to those skilled in the art. [0046]
  • “Specifically cleave”, when referring to the action of one of the instant catalytic deoxyribonucleic acid molecules on a target mRNA molecule, shall mean to cleave the target mRNA molecule without cleaving another mRNA molecule lacking a sequence complementary to either of the catalytic deoxyribonucleic acid molecule's two binding domains. [0047]
  • “Subject” shall mean any animal, such as a human, a primate, a mouse, a rat, a guinea pig or a rabbit. [0048]
  • The terms “DNA enzyme”, “DNAzyme” and “catalytic deoxyribonucleic acid” are used synonymously. [0049]
  • “Vector” shall include, without limitation, a nucleic acid molecule that can be used to stably introduce a specific nucleic acid sequence into the genome of an organism. [0050]
  • This invention provides a catalytic nucleic acid that specifically cleaves an mRNA encoding a Plasminogen Activator Inhibitor-1 (PAI-1) comprising, in 5′ to 3′ order: [0051]
  • (a) consecutive nucleotides defining a first binding domain of at least 4 nucleotides; [0052]
  • (b) consecutive nucleotides defining a catalytic domain located contiguous with the 3′ end of the first binding domain, and capable of cleaving the PAI-1-encoding mRNA at a predetermined phosphodiester bond; and [0053]
  • (c) consecutive nucleotides defining a second binding domain of at least 4 nucleotides located contiguous with the 3′ end of the catalytic domain, [0054]
  • wherein the sequence of the nucleotides in each binding domain is complementary to a sequence of ribonucleotides in the PAI-1-encoding mRNA and wherein the catalytic nucleic acid hybridizes to and specifically cleaves the PAI-1-encoding mRNA. [0055]
  • In different embodiments the catalytic deoxyribonucleic acid molecules can cleave the PAI-1-encoding mRNA at the linkage between 5′-A (or G) and U(T)-3′ (or C) i.e. at purine:pyrimidine consensus sequences. In further embodiments the catalytic deoxyribonucleic acid molecules cleaves the PAI-1 mRNA at the linkage of any 5′-AU-3′; 5′-AC-3′; 5′-GU-3′; or 5′-GC-3′ site within the mRNA. [0056]
  • To target the human PAI-1-encoding mRNA, catalytic nucleic acids can be designed based on the [0057] consensus cleavage sites 5′-purine:pyrimidine-3′ in the mRNA sequence (GenBank Accession#: M16006) (36). Those potential cleavage sites located on an open loop of the mRNA according to RNA folding software e.g. RNAdraw 2.1 are particularly preferred as targets (22). The DNA based catalytic nucleic acids can utilize the structure where two sequence-specific arms are attached to a catalytic core (SEQ ID NO:1) based on the PAI-1-encoding mRNA sequence (corresponding DNA shown as SEQ ID NO:5). Further examples of catalytic DNA structure are detailed in (23) and (24). Commercially available mouse brain polyA-RNA (Ambion) can serve as a template in the in vitro cleavage reaction to test the efficiency of the catalytic deoxyribonucleic acids.
  • Catalytic nucleic acid molecules can cleave PAI-1-encoding mRNA at each and any of the consensus sequences therein. Since catalytic ribo- and deoxyribo-nucleic acid consensus sequences are known, and the PAI-1-encoding mRNA sequence is known, one of ordinary skill could readily construct a catalytic ribo- or deoxyribo nucleic acid molecule directed to any of the PAI-1-encoding mRNA consensus sequences based on the instant specification. In preferred embodiments of this invention the catalytic deoxyribonucleic acids include the 10-23 structure. Cleavage of PAI-1 mRNA by DNAzyme may occur at 264 cleavage sites in the coding region of human PAI-1-encoding mRNA, including 51 5′-AT-3′ (-AU-) sites, 76 -AC- sites, 53 -GT- (-GU-) sites and 84 -GC- sites. See FIGS. 10 and 11 for AT and AC cleavage sites respectively, represented on the DNA sequence corresponding to the PAI-1-encoding mRNA. [0058]
  • Taking -AT- sites as a example, cleavage sites include [0059] nucleotide 76, 82, 152, 159, 254, 277, 316, 328, 333, 340, 344, 355, 374, 391, 399, 410, 415, 433, 449, 472, 543, 547, 550, 554, 583, 586, 644, 717, 745, 748, 773, 794, 800, 811, 847, 853, 866, 901, 919, 939, 1027, 1036, 1120, 1125, 1168, 1183, 1198, 1201, 1204, 1261, and 1273 of SEQ ID NO:5.
  • Examples of catalytic ribonucleic acids include hairpin and hammerhead ribozymes. In preferred embodiments of this invention, the catalytic ribonucleic acid molecule is formed in a hammerhead (25) or hairpin motif (26,27,28), but may also be formed in the motif of a hepatitis delta virus (29), group I intron (35), RNaseP RNA (in association with an RNA guide sequence) (30,31) or Neurospora VS RNA (32,33,34). Hammerhead ribozymes can cleave any 5′-NUH-3′ triplets of a mRNA, where U is conserved and N is any nucleotide and H can be C,U,A, but not G. For example, there are 151 sites which can be cleaved by a hammerhead ribozyme in human PAI-1-encoding mRNA coding region. See FIG. 12. [0060]
  • Cleaving of PAI-1-encoding mRNA with catalytic nucleic acids interferes with one or more of the normal functions of PAI-1-encoding mRNA. The functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA. [0061]
  • In one embodiment the nucleotides of the first binding domain comprise at least one deoxyribonucleotide. In another embodiment the nucleotides of the second binding domain comprise at least one deoxyribonucleotide. In one embodiment the nucleotides of the first binding domain comprise at least one deoxyribonucleotide derivative. In another embodiment the nucleotides of the second binding domain comprise at least one deoxyribonucleotide derivative. In one embodiment the nucleotides of the first binding domain comprise at least one ribonucleotide. In another embodiment the nucleotides of the second binding domain comprise at least one ribonucleotide. In one embodiment the nucleotides of the first binding domain comprise at least one ribonucleotide derivative. In another embodiment the nucleotides of the second binding domain comprise at least one ribonucleotide derivative. In one embodiment the nucleotides of the first binding domain comprise at least one modified base. In another embodiment the nucleotides of the second binding domain comprise at least one modified base. [0062]
  • The nucleotides may comprise other bases such as inosine, deoxyinosine, hypoxanthine may be used. In addition, [0063] isoteric purine 2′deoxy-furanoside analogs, 2′-deoxynebularine or 2′deoxyxanthosine, or other purine or pyrimidine analogs may also be used. By carefully selecting the bases and base analogs, one may fine tune the hybridization properties of the oligonucleotide. For example, inosine may be used to reduce hybridization specificity, while diaminopurines may be used to increase hybridization specificity.
  • Adenine and guanine may be modified at positions N3, N7, N9, C2, C4, C5, C6, or C8 and still maintain their hydrogen bonding abilities. Cytosine, thymine and uracil may be modified at positions N1, C2, C4, C5, or C6 and still maintain their hydrogen bonding abilities. Some base analogs have different hydrogen bonding attributes than the naturally occurring bases. For example, 2-amino-2′-dA forms three (3), instead of the usual two (2), hydrogen bonds to thymine (T). Examples of base analogs that have been shown to increase duplex stability include, but are not limited to, 5-fluoro-2′-dU, 5-bromo-2′-dU, 5-methyl-2′-dC, 5-propynyl-2′-dC, 5-propynyl-2′-dU, 2-amino-2′-dA, 7-deazaguanosine, 7-deazadenosine, and N2-Imidazoylpropyl-2′-dG. [0064]
  • Nucleotide analogs may be created by modifying and/or replacing a sugar moiety. The sugar moieties of the nucleotides may also be modified by the addition of one or more substituents. For example, one or more of the sugar moieties may contain one or more of the following substituents: amino, alkylamino, araalkyl, heteroalkyl, heterocycloalkyl, aminoalkylamino, O, H, an alkyl, polyalkylamino, substituted silyl, F, Cl, Br, CN, CF[0065] 3, OCF3, OCN, O-alkyl, S-alkyl, SOMe, SO2Me, ONO2, NH-alkyl, OCH2CH═CH2, OCH2CCH, OCCHO, allyl, O-allyl, NO2, N3, and NH2. For example, the 2′ position of the sugar may be modified to contain one of the following groups: H, OH, OCN, O-alkyl, F, CN, CF3, allyl, O-allyl, OCF3, S-alkyl, SOMe, SO2Me, ONO2, NO2, N3, NH2, NH-alkyl, or OCH═CH2, OCCH, wherein the alkyl may be straight, branched, saturated, or unsaturated. In addition, the nucleotide may have one or more of its sugars modified and/or replaced so as to be a ribose or hexose (i.e. glucose, galactose) or have one or more anomeric sugars. The nucleotide may also have one or more L-sugars.
  • Representative United States patents that teach the preparation of such modified bases/nucleosides/nucleotides include, but are not limited to, U.S. Pat. Nos. 6,248,878, and 6,251,666 which are herein incorporated by reference. [0066]
  • The sugar may be modified to contain one or more linkers for attachment to other chemicals such as fluorescent labels. In an embodiment, the sugar is linked to one or more aminoalkyloxy linkers. In another embodiment, the sugar contains one or more alkylamino linkers. Aminoalkyloxy and alkylamino linkers may be attached to biotin, cholic acid, fluorescein, or other chemical moieties through their amino group. [0067]
  • Nucleotide analogs or derivatives may have pendant groups attached. Pendant groups serve a variety of purposes which include, but are not limited to, increasing cellular uptake of the molecule, enhancing degradation of the target nucleic acid, and increasing hybridization affinity. Pendant groups can be linked to the binding domains of the catalytic nucleic acid. Examples of pendant groups include, but are not limited to: acridine derivatives (i.e. 2-methoxy-6-chloro-9-aminoacridine); cross-linkers such as psoralen derivatives, azidophenacyl, proflavin, and azidoproflavin; artificial endonucleases; metal complexes such as EDTA-Fe(II), o-phenanthroline-Cu(I), and porphyrin-Fe(II); alkylating moieties; nucleases such as amino-1-hexanolstaphylococcal nuclease and alkaline phosphatase; terminal transferases; abzymes; cholesteryl moieties; lipophilic carriers; peptide conjugates; long chain alcohols; phosphate esters; amino; mercapto groups; radioactive markers; nonradioactive markers such as dyes; and polylysine or other polyamines. In one example, the nucleic acid comprises an oligonucleotide conjugated to a carbohydrate, sulfated carbohydrate, or gylcan. Conjugates may be regarded as a way as to introduce a specificity into otherwise unspecific DNA binding molecules by covalently linking them to a selectively hybridizing oligonucleotide. [0068]
  • The binding domains of the catalytic nucleic acid may have one or more of their sugars modified or replaced so as to be ribose, glucose, sucrose, or galactose, or any other sugar. Alternatively, they may have one or more sugars substituted or modified in its 2′ position, i.e. 2′allyl or 2′-O-allyl. An example of a 2′-O-allyl sugar is a 2′-O-methylribonucleotide. Further, the nucleotides of the binding domain may have one or more of their sugars substituted or modified to form an α-anomeric sugar. [0069]
  • A catalytic nucleic acid binding domain may include non-nucleotide substitution. The non-nucleotide substitution includes either abasic nucleotide, polyether, polyamine, polyamide, peptide, carbohydrate, lipid or polyhydrocarbon compounds. The term “abasic” or “abasic nucleotide” as used herein encompasses sugar moieties lacking a base or having other chemical groups in place of base at the 1′ position. [0070]
  • In one embodiment the nucleotides of the first binding domain comprise at least one modified internucleoside bond. In another embodiment the nucleotides of the second binding domain comprise at least one modified internucleoside bond. In a further embodiment the modified internucleoside bond is a phosphorothioate bond. The nucleic acid may comprise modified bonds. For example the bonds between nucleotides of the catalytic nucleic acid may comprise phosphorothioate linkages. The nucleic acid may comprise nucleotides having moiety may be modified by replacing one or both of the two bridging oxygen atoms of the linkage with analogues such as —NH, —CH[0071] 2, or —S. Other oxygen analogues known in the art may also be used. The phosphorothioate bonds may be stereo regular or stereo random.
  • In one embodiment the PAI-1-encoding mRNA encodes human PAI-1. In a further embodiment the human PAI-1-encoding mRNA has the sequence set forth in SEQ ID NO:5. [0072]
  • This invention provides a pharmaceutical composition comprising the instant catalytic nucleic acid and a pharmaceutically acceptable carrier. The following pharmaceutically acceptable carriers are set forth, in relation to their most commonly associated delivery systems, by way of example, notwithstanding the fact that the instant pharmaceutical compositions are preferably delivered dermally. [0073]
  • Dermal delivery systems include, for example, aqueous and nonaqueous gels, creams, multiple emulsions, microemulsions, liposomes, ointments, aqueous and nonaqueous solutions, lotions, aerosols, hydrocarbon bases and powders, and can contain excipients such as solubilizers, permeation enhancers (e.g., fatty acids, fatty acid esters, fatty alcohols and amino acids), and hydrophilic polymers (e.g., polycarbophil and polyvinylpyrolidone). In one embodiment, the pharmaceutically acceptable carrier is a liposome or a transdermal enhancer. Examples of liposomes which can be used in this invention include the following: (1) CellFectin, 1:1.5 (M/M) liposome formulation of the cationic lipid N,NI,NII,NIII-tetramethyl-N,NI,NII,NIII-tetrapalmity-spermine and dioleoyl phosphatidylethanolamine (DOPE)(GIBCO BRL); (2) Cytofectin GSV, 2:1 (M/M) liposome formulation of a cationic lipid and DOPE (Glen Research); (3) DOTAP (N-[1-(2,3-dioleoyloxy)-N,N,N-tri-methyl-ammoniumethyl sulfate) (Boehringer Manheim); and (4) Lipofectamine, 3:1 (M/M) liposome formulation of the polycationic lipid DOSPA and the neutral lipid DOPE (GIBCO BRL). [0074]
  • Transmucosal delivery systems include patches, tablets, suppositories, pessaries, gels and creams, and can contain excipients such as solubilizers and enhancers (e.g., propylene glycol, bile salts and amino acids), and other vehicles (e.g., polyethylene glycol, fatty acid esters and derivatives, and hydrophilic polymers such as hydroxypropylmethylcellulose and hyaluronic acid). [0075]
  • Injectable drug delivery systems include solutions, suspensions, gels, microspheres and polymeric injectables, and can comprise excipients such as solubility-altering agents (e.g., ethanol, propylene glycol and sucrose) and polymers (e.g., polycaprylactones and PLGA's). Implantable systems include rods and discs, and can contain excipients such as PLGA and polycaprylactone. [0076]
  • Oral delivery systems include tablets and capsules. These can contain excipients such as binders (e.g., hydroxypropylmethylcellulose, polyvinyl pyrilodone, other cellulosic materials and starch), diluents (e.g., lactose and other sugars, starch, dicalcium phosphate and cellulosic materials), disintegrating agents (e.g., starch polymers and cellulosic materials) and lubricating agents (e.g., stearates and talc). [0077]
  • This invention provides a method of specifically inhibiting the expression of PAI-1 in a cell that would otherwise express PAI-1, comprising contacting the cell with the instant catalytic nucleic acid so as to specifically inhibit the expression of PAI-1 in the cell. [0078]
  • This invention provides a method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the instant catalytic nucleic acid effective to specifically inhibit the expression of PAI-1 in the subject's cells. [0079]
  • This invention provides a method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the instant pharmaceutical composition effective to specifically inhibit the expression of PAI-1 in the subject's cells. [0080]
  • This invention provides a method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the instant pharmaceutical composition effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease. [0081]
  • In one embodiment the cardiovascular disease is congestive heart failure. In another embodiment the cardiovascular disease is myocardial infarct. In another embodiment the cardiovascular disease is angina. In another embodiment the cardiovascular disease is myocardial ischemia. In another embodiment the cardiovascular disease is a cardiomyopathy. [0082]
  • This invention provides a method of treating a fibrotic disease in a subject involving fibrogenesis which comprises administering to the subject an amount of the instant pharmaceutical composition effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease. In differing embodiments the fibrotic disease is a renal disease, a hepatic disease, a disease of the lung, a disease of the skin, or a disease of the eye. In further embodiments the fibrotic disease of the skin is scleroderma or psorasis. [0083]
  • An “effective amount” is an amount at least sufficient to treat, reduce, reverse or otherwise inhibit the given disease state. In the case of fibrotic diseases this means an amount sufficient to reduce, reverse or otherwise inhibit fibrogenesis. In the case of cardiovascular diseases this means an amount sufficient to reduce, reverse or otherwise inhibit cardiomyocyte apoptosis. [0084]
  • Determining the effective amount of the instant pharmaceutical compositions can be done based on animal data using routine computational methods. In one embodiment, the effective amount contains between about 10 ng and about 100 μg of the instant nucleic acid molecules per kg body mass. In another embodiment, the effective amount contains between about 100 ng and about 10 μg of the nucleic acid molecules per kg body mass. In a further embodiment, the effective amount contains between about 1 μg and about 5 μg of the nucleic acid molecules per kg body mass. In another embodiment the effective amount contains between about 10 μg and about 100 μg of the nucleic acid molecules per kg body mass. In a preferred embodiment the effective amount contains between about 100 μg and 500 μg of the nucleic acid molecules per kg body mass. In another embodiment the effective amount contains between about 500 μg and 1000 μg of the nucleic acid molecules per kg body mass. In another embodiment the effective amount contains between about 1 mg and 2 mg of the nucleic acid molecules per kg body mass. [0085]
  • The compounds of the invention may also be admixed, encapsulated, conjugated or otherwise associated with other molecules, molecule structures or mixtures of compounds, as for example, liposomes, receptor targeted molecules, oral, rectal, topical or other formulations, for assisting in uptake, distribution and/or absorption. Representative United States patents that teach the preparation of such uptake, distribution and/or absorption assisting formulations include, but are not limited to, U.S. Pat. Nos. 5,108,921; 5,354,844; 5,416,016; 5,459,127; 5,521,291; 5,543,158; 5,547,932; 5,583,020; 5,591,721; 4,426,330; 4,534,899; 5,013,556; 5,108,921; 5,213,804; 5,227,170; 5,264,221; 5,356,633; 5,395,619; 5,416,016; 5,417,978; 5,462,854; 5,469,854; 5,512,295; 5,527,528; 5,534,259; 5,543,152; 5,556,948; 5,580,575; and 5,595,756, each of which is herein incorporated by reference. [0086]
  • It is preferred to administer catalytic nucleic acids to mammals suffering from a cardiovascular disease or a fibrotic disease, in either native form or suspended in a carrier medium in amounts and upon treatment schedules which are effective to therapeutically treat the mammals to reduce the detrimental effects of cardiovascular disease or fibrotic disease. One or more different catalytic nucleic acids or antisense oligonucleotides or analogs thereof targeting different sections of the nucleic acid sequence of PAI-1-encoding mRNA may be administered together in a single dose or in different doses and at different amounts and times depending upon the desired therapy. The catalytic nucleic acids can be administered to mammals in a manner capable of getting the nucleic acids initially into the blood stream and subsequently into cells, or alternatively in a manner so as to directly introduce the catalytic nucleic acids into the cells or groups of cells, for example cardiomyocytes, by such means by electroporation or by direct injection into the heart or by catheter into renal tissue. It is within the scale of a person's skill in the art to determine optimum dosages and treatment schedules for such treatment regimens. [0087]
  • The effective amount of catalytic nucleic acid is administered to a human patient in need of inhibition of PAI-1 expression (or inhibition of fibrogenesis) from 1-8 or more times daily or every other day. Dosage is dependent on severity and responsiveness of the effects of the cardiovascular or fibrotic disease to be treated, with a course of treatment lasting from several days to months or until a cure is effected or a reduction of the effects is achieved. The actual effective amount, or dosage, administered may take into account the size and weight of the patient, whether the nature of the treatment is prophylactic, therapeutic in nature, the age, weight, health and sex of the patient, the route of administration, and other factors. [0088]
  • Pharmaceutical compositions may contain suitable excipients and auxiliaries which facilitate processing of the catalytic nucleic acids into preparations which can be used pharmaceutically. Preferably, the preparations, particularly those which can be administered orally and which can be used for the preferred type of administration, such as tablets, dragees, and capsules, and preparations which can be administered rectally, such as suppositories, as well as suitable solutions for administration parenterally or orally, and compositions which can be administered bucally or sublingually, including inclusion compounds, contain from about 0.1 to about 99 percent by weight of active ingredients, together with the excipient. [0089]
  • The pharmaceutical preparations of the present invention are manufactured in a manner which is itself well known in the art. For example, the pharmaceutical preparations may be made by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes. The process to be used will depend ultimately on the physical properties of the active ingredient used. [0090]
  • Suitable excipients are, in particular, fillers such as sugars, for example, lactose or sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example, tricalcium phosphate or calcium hydrogen phosphate as well as binders such as starch, paste, using, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone. If desired, disintegrating agents may be added, such as the above-mentioned starches as well as carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate. Auxiliaries are flow-regulating agents and lubricants, for example, such as silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol. Dragee cores may be provided with suitable coatings which, if desired, may be resistant to gastric juices. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinylpyrrolidone, polyethylene, glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. In order to produce coatings resistant to gastric juices, solutions of suitable cellulose preparations such as acetyl-cellulose phthalate or hydroxypropylmethycellulose phthalate, are used. Dyestuffs and pigments may be added to the tablets of dragee coatings, for example, for identification or in order to characterize different combinations of active compound doses. Other pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. The push-fit capsules can contain the active compounds in the form of granules which may be mixed with filters such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and optionally, stabilizers. In soft capsules, the active compounds are preferably dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In additions, stabilizers may be added. [0091]
  • Possible pharmaceutical preparations which can be used rectally include, for example, suppositories, which consist of a combination of the active compounds with a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, paraffin hydrocarbons, polyethylene glycols or higher alkanols. In addition, it is also possible to use gelatin rectal capsules which consist of a combination of the active compounds with a base. Possible base materials include, for example liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons. [0092]
  • Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble or water-dispersible form. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, ethyl oleate or triglycerides. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers. [0093]
  • Additionally, catalytic nucleic acids of the present invention may also be administered encapsulated in liposomes or immunoliposomes, which are pharmaceutical compositions wherein the active ingredient is contained either dispersed or variously present in corpuscles consisting of aqueous concentric layers adherent to lipidic layers. Liposomes are especially active in targeting the oligonucleotides to liver cells. The active ingredient, depending upon its solubility, may be present both in the aqueous layer and in the lipidic layer, or in what is generally termed a liposomic suspension. The hydrophobic layer, generally but not exclusively, comprises phospholipids such as lecithin and sphingomyelin, steroids such as cholesterol, more or less ionic surfactants such as dicetylphosphate, stearylamine, or phosphatidic acid, and/or other materials of a hydrophobic nature. The diameters of the liposomes generally range from about 15 nm to about 5 microns. [0094]
  • This invention also provides an oligonucleotide comprising consecutive nucleotides that hybridizes with a PAI-1-encoding mRNA under conditions of high stringency and is between 8 and 40 nucleotides in length. In different embodiments the oligonucleotide is between 40 and 80 nucleotides in length. [0095]
  • In a preferred embodiment the hybridization of antisense oligonucleotides with PAI-1-encoding mRNA interferes with one or more of the normal functions of PAI-1-encoding mRNA. The functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA. For example, a human PAI-1 antisense oligonucleotide specifically hybridizes under given stringent conditions with targets on the human PAI-1 mRNA molecule and in doing so inhibits the translation thereof into PAI-1 protein. [0096]
  • The functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA. A PAI-1 antisense oligonucleotide specifically hybridizes under given stringent conditions with targets on the PAI-1-encoding mRNA molecule and in doing so inhibits the translation thereof into PAI-1. [0097]
  • In accordance with this invention, persons of ordinary skill in the art will understand that messenger RNA includes not only the information to encode a protein using the three letter genetic code, but also associated ribonucleotides which form a region known to such persons as the 5′-untranslated region, the 3′-untranslated region, the 5′ cap region and intron/exon junction ribonucleotides. Thus, catalytic nucleic acids or antisense oligonucleotides may be formulated in accordance with this invention which are targeted wholly or in part to these associated ribonucleotides as well as to the informational ribonucleotides. For example, the antisense oligonucleotides may therefore be specifically hybridizable with a transcription initiation site region, a translation initiation codon region, a 5′ cap region, an intron/exon junction, coding sequences, a translation termination codon region or sequences in the 5′- or 3′-untranslated region. Similarly, the catalytic nucleic acids may specifically cleave a transcription initiation site region, a translation initiation codon region, a 5′ cap region, an intron/exon junction, coding sequences, a translation termination codon region or sequences in the 5′- or 3′-untranslated region. As is known in the art, the translation initiation codon is typically 5′-AUG (in transcribed mRNA molecules; 5′-ATG in the corresponding DNA molecule). A minority of genes have a translation initiation codon having the [0098] RNA sequence 5′-GUG, 5′UUG or 5′-CUG, and 5′-AUA, 5′-ACG and 5′-CUG have been shown to function in vivo. Thus, the term “translation initiation codon” can encompass many codon sequences, even though the initiator amino acid in each instance is typically methionine in eukaryotes. It is also known in the art that eukaryotic genes may have two or more alternative translation initiation codons, any one of which may be preferentially utilized for translation initiation in a particular cell type or tissue, or under a particular set of conditions. In the context of the invention, “translation initiation codon” refers to the codon or codons that are used in vivo to initiate translation of an mRNA molecule transcribed from a gene encoding PAI-1, regardless of the sequence(s) of such codons. It is also known in the art that a translation termination codon of a gene may have one of three sequences, i.e., 5′-UAA, 5′-UAG and 5′-UGA (the corresponding DNA sequences are 5′-TAA, 5′-TAG and 5′-TGA, respectively). The term “translation initiation codon region” refers to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′) from a translation initiation codon. This region is one preferred target region. Similarly, the term “translation termination codon region” refers to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′) from a translation termination codon. This region is also one preferred target region. The open reading frame or “coding region,” which is known in the art to refer to the region between the translation initiation codon and the translation termination codon, is also a region which may be targeted effectively. Other preferred target regions include the 5′ untranslated region (5′UTR), known in the art to refer to the portion of an mRNA in the 5′ direction from the translation initiation codon, and thus including nucleotides between the 5′ cap site and the translation initiation codon of an mRNA or corresponding nucleotides on the gene, and the 3′ untranslated region (3′UTR), known in the art to refer to the portion of an mRNA in the 3′ direction from the translation termination codon, and thus including nucleotides between the translation termination codon and 3′ end of an mRNA or corresponding nucleotides on the gene. mRNA splice sites may also be preferred target regions, and are particularly useful in situations where aberrant splicing is implicated in disease, or where an overproduction of a particular mRNA splice product is implicated in disease. Aberrant fusion junctions due to rearrangements or deletions may also be preferred targets.
  • Once the target site or sites have been identified, antisense oligonucleotides can be chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired disruption of the function of the molecule. “Hybridization”, in the context of this invention, means hydrogen bonding, also known as Watson-Crick base pairing, between complementary bases, usually on opposite nucleic acid strands or two regions of a nucleic acid strand. Guanine and cytosine are examples of complementary bases which are known to form three hydrogen bonds between them. Adenine and thymine are examples of complementary bases which form two hydrogen bonds between them. “Specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of complementarity such that stable and specific binding occurs between the DNA or RNA target and the antisense oligonucleotide. Similarly, catalytic nucleic acids are synthesized once cleavage target sites on the PAI-1 mRNA molecule have been identified, e.g. any purine:pyrimidine consensus sequences in the case of DNA enzymes. Catalytic nucleic acids and antisense oligonucleotides targeted to disrupting polymorphisms and mutants of the PAI-1 maybe similarly made as described above based on the polymorphic or mutant PAI-1 mRNA sequence. [0099]
  • Methods for selecting which particular antisense oligonucleotides sequences directed towards a particular protein-encoding mRNA are that will form the most stable DNA:RNA hybrids within the given target mRNA sequence are known in the art and are exemplified in U.S. Pat. No. 6,183,966 which is herein incorporated by reference. [0100]
  • In one embodiment at least one internucleoside linkage within the instant oligonucleotide comprises a phosphorothioate linkage. Antisense oligonucleotide molecules synthesized with a phosphorothioate backbone have proven particularly resistant to exonuclease damage compared to standard deoxyribonucleic acids, and so they are used in preference. A phosphorothioate antisense oligonucleotide for PAI-1 mRNA can be synthesized on an Applied Biosystems (Foster City, Calif.) model 380B DNA synthesizer by standard methods. For example, sulfurization can be performed using tetraethylthiuram disulfide/acetonitrile. Following cleavage from controlled pore glass support, oligodeoxynucleotides can be base deblocked in ammonium hydroxide at 60° C. for 8 h and purified by reversed-phase HPLC [0.1M triethylammonium bicarbonate/acetonitrile; PRP-1 support]. Oligomers can be detritylated in 3% acetic acid and precipitated with 2% lithiumperchlorate/acetone, dissolved in sterile water and reprecipitated as the sodium salt from 1 M NaCl/ethanol. Concentrations of the full length species can be determined by UV spectroscopy. Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and alkylated derivatives. [0101]
  • Preferred modified oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3T-alkylene phosphonates, 5′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and borano-phosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3′ to 3′, 5′ to 5′ or 2′ to 2′ linkage. Preferred oligonucleotides having inverted polarity comprise a single 3′ to 3′ linkage at the 3′-most internucleotide linkage i.e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof). Various salts, mixed salts and free acid forms are also included. Representative United States patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,194,599; 5,565,555; 5,527,899; 5,721,218; 5,672,697 and 5,625,050, each of which is herein incorporated by reference. [0102]
  • In one embodiment the nucleotides of the instant oligonucleoptide comprise at least one deoxyribonucleotide. In another embodiment the nucleotides comprise at least one ribonucleotide. Such deoxyribonucleotides or ribonucleotides can be modified or derivatized as described hereinabove. [0103]
  • In one embodiment the PAI-1-encoding mRNA encodes human PAI-1. In a further embodiment the human PAI-1-encoding mRNA comprises consecutive nucleotides, the sequence of which is set forth in SEQ ID NO:5. [0104]
  • This invention further provides a method of treating a subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit expression of a PAI-1 in the subject so as to thereby treat the subject. [0105]
  • This invention further provides a method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease. [0106]
  • This invention further provides a method of treating a fibrotic disease in a subject involving fibrogenesis in the subject which comprises administering to the subject an amount of the instant oligonucleotide effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease. [0107]
  • Antisense oligonucleotides can be administered by intravenous injection, intravenous drip, subcutaneous, intraperitoneal or intramuscular injection, orally or rectally. Human pharmacokinetics of certain antisense oligonucleotides have been studied. See (37) incorporated by reference in its entirety. [0108]
  • This invention provides all of the instant methods wherein the subject is a mammal. In a preferred embodiment the mammal is a human being. [0109]
  • In this invention, the various embodiments of subjects, pharmaceutically acceptable carriers, dosage, cell types, routes of administration and target nucleic acid sequences are envisioned for each of the instant nucleic acid molecules, oligonucleotides, pharmaceutical compositions and methods. [0110]
  • This invention will be better understood by reference to the Experimental Details which follow, but those skilled in the art will readily appreciate that the specific experiments detailed are only illustrative of the invention as described more fully in the claims which follow thereafter. [0111]
  • Experimental Results [0112]
  • Construction of DNA Enzymes Targeting Human and Rat PAI-1 mRNA. [0113]
  • We designed three DNA enzymes to specifically target pyrimidine-purine junctions at or near the translational start site AUG of human and rat PAI-1 messenger RNA, a region that is conserved between species and has low relative free energy (17). As shown in FIG. 1[0114] a, the three DNA enzymes, termed E1 (SEQ ID NO:2), E2 (SEQ ID NO:4), and E3 (SEQ ID NO:3), contained identical 15-nucleotide catalytic domains (SEQ ID NO:1) which were flanked by two arms of eight nucleotides (E1, E2) or nine nucleotides (E3) with complementarity to human (E1, E3) or rat (E2) PAI-1 mRNA. To produce the control DNA enzyme E0 (SEQ ID NO:7), the nucleotide sequence in the two flanking arms of E2 was scrambled without altering the catalytic domain (FIG. 1a). The 3′ terminus of each molecule was capped with an inverted 3′-3′-linked thymidine for resistance to 3′-to-5′ exonuclease digestion.
  • Specific Cleavage of Human PAI-1 mRNA by E1 and E3 DNA Enzymes. [0115]
  • As shown in FIG. 1[0116] b, the 21-base oligonucleotide S1 (SEQ ID NO:8), synthesized from human PAI-1 mRNA and labeled at the 5′ end with 32P, was cleaved within 2 minutes when cultured with E1 at 10:1 substrate:enzyme excess, with maximal cleavage occurring by 2 hours. The 10-nucleotide cleavage product is consistent with the size of the 32P-labeled fragment at the 5′ end. E1 also cleaved larger 32P-labeled fragments of human PAI-1 mRNA, prepared by in vitro transcription, in a time- and concentration-dependent manner, FIG. 1c. A 520 nucleotide transcript was maximally cleaved by 2-4 hours to expected cleavage products of 320 and 200 nucleotides. A similar dose-dependency was seen with E3, with maximal cleavage of human PAI-1 mRNA transcripts occurring at the highest concentration used, 20 μM. The sequence-specific nature of the DNA enzymatic cleavage is shown in FIG. 1d, where the control DNA enzyme E0, containing an identical catalytic domain to E1 and E3, but scrambled sequences in the flanking arms, caused no cleavage of human PAI-1 mRNA transcripts. Pre-heating the transcript to 72° C. for 10 minutes prior to incubation with E1, but not E0, further increased cleavage.
  • Specific Cleavage of Rat PAI-1 mRNA by E2 DNA Enzyme. [0117]
  • Further evidence for the specificity of the DNA enzymatic reactions can be seen in FIG. 2. The DNA enzyme E2 cleaved the 23-base oligonucleotide S2, synthesized from the sequence of rat PAI-1 mRNA, in a dose- and time-dependent manner, FIG. 2[0118] a. E2 also cleaved a rat PAI-1 mRNA transcript in a dose-dependent manner by 2-4 hours to give the 156 nucleotide cleavage product, FIG. 2b. In contrast, neither the scrambled control DNA enzyme E0 nor E3 cleaved the rat PAI-1 mRNA transcript. Since the rat PAI-1 mRNA transcript (SEQ ID NO:10) differs by only one nucleotide from the human mRNA PAI-1 transcript (SEQ ID NO:9) which can be cleaved by E3, FIG. 2c, these results demonstrate the exquisite target specificity of these DNA enzymes.
  • DNA Enzymes Inhibit Induction of Endogenous PAI-1 mRNA and Protein. [0119]
  • To determine the effect of DNA enzymes on endogenous PAI-1 production, endothelial monolayers of human and rat origin were grown to confluence and transfected with species-specific DNA enzymes or scrambled control. Transfected cells were then activated with TGF-beta for 8 hours to maximally induce expression of PAI-1. Densitometric analysis of RT-PCR products following reverse transcription of cellular mRNA showed that E2 inhibited TGF-beta inducible steady-state mRNA levels in cultured rat endothelium by 52%, FIG. 3[0120] a, relative to the E0 scrambled DNA. Constitutive PAI-1 mRNA levels were not affected by transfection with the DNA enzymes. FIG. 3b shows the effect of endothelial cell transfection with E2 DNA enzyme on TGF-beta mediated induction of PAI-1 protein. Endothelial cells transfected with scrambled DNA enzyme demonstrated approximately 50% increase in cytoplasmic PAI-1 protein as detected by Western blot. In contrast, this effect was almost completely is abrogated by transfection with the PAI-1 DNA enzyme E2.
  • Resistance of DNA Enzymes to Serum-Dependent Degradation. [0121]
  • Since DNA enzymes with a thymidine in the correct orientation at the 3′ end are significantly degraded by factors in serum (1-5% concentration) within 24 hours (14), we investigated the protective effect of an inverted thymidine at the 3′ end on serum-dependent nucleolytic degradation of PAI-1 DNA enzymes. The DNA enzyme E1 was labeled with 32P and incubated for 3-24 hours with human umbilical vein endothelial cell (HUVEC) monolayers cultured in medium containing physiologic (2%) or supraphysiologic (20%) serum concentrations As shown in FIG. 3[0122] c, whereas the DNA enzyme was almost completely degraded within 6 hours of cell culture in medium containing 20% serum, it remained intact during the entire 24 hours of cell culture with medium containing 2% serum. As shown in FIG. 3d, HUVEC transfected with E1 or E3 DNA enzymes and cultured for 12 hours in 2% serum demonstrated 20% and 28% reduction in TGF-beta dependent PAI-1 activity compared with transfection with the scrambled DNA E0 (both p<0.01). Together, these results indicate that an inverted thymidine at the 3′ end can protect the PAI-1 DNA enzyme against nucleolytic degradation at physiologic serum concentrations likely to be encountered in vivo, consequently enabling inhibition of cellular PAI-1 activity.
  • Intracardiac Injection of E2 Reduces Macrophage PAI-1 Expression. [0123]
  • We investigated whether intracardiac injection of E2 at the time of myocardial infarction could inhibit PAI-1 expression in the rat heart. At two weeks after ligation of the left anterior descending (LAD) artery and induction of myocardial infarction, immunohistochemical studies showed that PAI-1 expression occurred predominantly in CD68-positive rat macrophages within the infarct zone and at the peri-infarct region. While a few vascular endothelial cells stained positively, PAI-1 expression was not detected within myocytes or interstitial cells distally to the infarct. We conclude that the principal source of PAI-1 protein in the infarcted rat heart is due to production by infiltrating macrophages and phagocytes. At two weeks after injection with E2, PAI-1 expression in infarcted rat hearts was dramatically altered, with 71% reduction in PAI-1 expression by macrophages at the peri-infarct region (p<0.01), FIG. 4[0124] a. Inhibition of PAI-1 expression at this site was highly specific since PAI-1 expression within macrophages in the center of the infarct zone remained unaffected after E2 injection, and injection with the scrambled DNA enzyme E0 did not reduce peri-infarct PAI-1 expression.
  • Intracardiac Injection of E2 Results in Peri-Infarct Neovascularization. [0125]
  • By two weeks, in parallel with reduction in PAI-1 expression, hearts from rats injected with E2 demonstrated significantly increased numbers of large-lumen capillaries (>6 nuclei) at the peri-infarct region, FIG. 4[0126] b. While injection of E2 alone caused a significant increase in large-lumen capillaries at the peri-infarct region compared with injection of the E0 scrambled DNA enzyme, even more striking was the additional 62% increase in the number of large-lumen capillaries when E2 was co-administered together with intravenously delivered human adult bone marrow-derived CD34+CD117bright angioblasts. While many of the newly-formed vessels were of rat origin (angiogenesis), a significant number were of human origin (vasculogenesis), as defined by a human anti-CD31 mAb. In the presence of E2 the number of unincorporated, discrete, interstitial human angioblasts in the infarct zone and peri-infarct region was reduced by 5.2-fold compared with E0 control enzyme, FIG. 4c. Conversely, the number of large-lumen capillaries (>6 nuclei) at the peri-infarct region was increased by 14-fold following E2 injection as compared with E0. We conclude that by inhibiting PAI-1 expression at the peri-infarct region E2 enables human angioblasts to more effectively migrate through cardiac tissue, coalesce and participate in new vessel formation.
  • E2 Protects Cardiomyocytes at the Peri-Infarct Region Against Apoptosis. [0127]
  • We have previously shown that neovascularization at the peri-infarct region, induced by bone marrow-derived endothelial progenitors, can protect adjacent viable cardiomyocytes against apoptosis. Since intracardiac injection of E2 alone induced neovascularization at this site in infarcted rat hearts, we examined the effect of E2 injection on cardiomyocyte apoptosis. Injection with E2 at the time of LAD ligation resulted in 70% reduction in apoptosis of cardiomyocytes at the peri-infarct region (p<0.01), FIG. 5[0128] a. This was not observed with injection of the scrambled control DNA enzyme E0. The level in protection against cardiomyocyte apoptosis was similar to the level observed with intravenous infusion of human angioblasts.
  • Renal Fibrosis. [0129]
  • The renin-angiotensin system (RAS) in progressive renal disease has been extensively investigated, indicating multiple actions beyond hemodynamic and salt/water homeostasis. The RAS is now recognized to be linked to induction of plasminogen activator inhibitor-1 (PAI-1) likely via both the type 1 (AT1) and type 4 (AT4) receptors, thus, promoting both thrombosis and fibrosis (39). Plasminogen [0130] activator inhibitor type 1 is thus a suitable target in renal fibrogenesis. The progression of renal lesions to fibrosis involves several mechanisms, among which the inhibition of extracellular matrix (ECM) degradation appears to play an important role. Two interrelated proteolytic systems are involved in matrix degradation: the plasminogen activation system and the matrix metalloproteinase system. PAI-1 as the main inhibitor of plasminogen activation, regulates fibrinolysis and the plasmin-mediated matrix metalloproteinase activation. PAI-1 is also a component of the ECM, where it binds to vitronectin. PAI-1 is not expressed in the normal human kidney but is strongly induced in various forms of kidney diseases, leading to renal fibrosis and terminal renal failure. Thrombin, angiotensin II, and transforming growth factor-beta are potent in vitro and in vivo agonists in increasing PAI-1 synthesis. Several experimental and clinical studies support a role for PAI-1 in the renal fibrogenic process occurring in chronic glomerulonephritis, diabetic nephropathy, focal segmental glomerulosclerosis, and other fibrotic renal diseases (40).
  • End-stage renal disease (ESRD) comprises an enormous public health burden, with an incidence and prevalence that are increasingly on the rise. This escalating prevalence suggests that newer therapeutic interventions and strategies are needed to complement current therapeutic approaches. Although much evidence demonstrates conclusively that angiotensin II mediates progressive renal disease, recent evidence also implicates aldosterone as an important pathogenetic factor in progressive renal disease. Recently, several lines of experimental evidence demonstrate that selective blockade of aldosterone, independent of renin-angiotensin blockade, reduces proteinuria and nephrosclerosis in the spontaneously hypertensive stroke-prone rat (SHRSP) model and reduces proteinuria and glomerulosclerosis in the subtotally nephrectomized rat model (ie, remnant kidney). Whereas pharmacologic blockade with angiotensin II receptor blockers and angiotensin-converting enzyme (ACE) inhibitors reduces proteinuria and nephrosclerosis/glomerulosclerosis, selective reinfusion of aldosterone restores these abnormalities despite continued renin-angiotensin blockade. Aldosterone may promote fibrosis by several mechanisms, including plasminogen activator inhibitor-1 (PAI-1) expression and consequent alterations of vascular ribrinolysis, by stimulation of transforming growth factor-betal (TGF-betal), and by stimulation of reactive oxygen species (ROS) (41). Therefore it is expected that PAI-1 is a suitable target for therapeutic intervention into renal fibrosis. [0131]
  • Progressive renal disease is characterized by the induction of plasminogen activator inhibitor-1 (PAI-1), suggesting that impaired activity of the renal plasmin cascade may play a role in renal fibrosis. Studies with PAI-1 knock-out mice showing resistance to fibrotic effects of ureteric obstruction establish an important fibrogenic role for PAI-1 in the renal fibrogenic response. The results demonstrate that one important fibrosis-promoting function of PAI-1 is its role in the recruitment of fibrosis-inducing cells, including myofibroblasts and macrophages (42). [0132]
  • Liver Fibrosis. [0133]
  • During chronic liver injury, transforming growth factor beta (TGF-beta) plays a prominent role in stimulating liver fibrogenesis by myofibroblast-like cells derived from hepatic stellate cells (HSCs). In acute liver injury, HSC-derived TGF-beta increased plasminogen activator inhibitor type 1 (PAI-1) and alpha2(I) procollagen (COL1A2) transcripts. [0134] Smad 2 in HSCs during liver injury and primary cultured HSCs were activated by an autocrine mechanism, because high levels of Smad 2 phosphorylation and induction of PAI-1 transcript by TGF-beta were observed in HSCs (43).
  • Increased PAI-1 together with uPA, uPAR and tPA are associated with overall inhibition of matrix degradation in cirrhotic liver. Hepatic stellate cells are an important source of PAI-1 during liver fibrosis. Increased expression of plasminogen activator and plasminogen activator inhibitor during liver fibrogenesis of rats: role of stellate cells (44). Therefore it is expected that PAI-1 is a suitable target for therapeutic intervention into hepatic fibrosis. [0135]
  • Lung Fibrosis: [0136]
  • PAI-1 levels in bronchoalveolar lavage (BAL) supernatant fluids are significantly higher in idiopathic pulmonary fibrosis (IPF) patients than in normal subjects. Increased procoagulant and antifibrinolytic activities in the lungs with idiopathic pulmonary fibrosis. Therefore it is expected that PAI-1 is a suitable target for therapeutic intervention into fibrosis of the lung. [0137]
  • In addition, cataracts of the eye may be treated by reversing the symptomatic fibrosis that occurs, thought to be due to ischemia, by the mechanism described hereinabove. [0138]
  • Materials and Methods [0139]
  • DNA enzymes and RNA substrates: DNA enzymes with 3′-3′ inverted thymidine were synthesized by Integrated DNA technologies (Coralville, Iowa) and purified by RNase-free IE-HPLC or RP-HPLC. The short RNA substrates corresponding to target DNA enzyme sequences were chemically synthesized followed by RNAse-free PAGE purification and also made by in vitro transcription from a DNA template. [0140]
  • In vitro transcription: Human PAI-1 cDNA was amplified by RT-PCR from total RNA of cultured HUVEC using the following primer pair: 5′CCAAGAGCGCTGTCAAGAAGAC3′ (forward primer; SEQ ID NO:11) and 5′TCACCGTCTGCTTTGGAGACCT3′ (reverse primer; SEQ ID NO:12) (position 25-10600, J. Biol. Chem, 1988, 263(19): 9143). Length of PCR product is 1598 bases. PAI-1 cDNA was cloned into pGEM-T vector (Promega) to obtain plasmid construct pGEM-hPAI. cDNA sequence was verified using an automatic sequencing machine. A 32P-labeled-nucleotide human PAI-1 RNA transcript was prepared by in vitro transcription (SP6 polymerase, Promega). 20 ml transcription reaction consisted of 4 [0141] μl 5× buffer, 2 μl DTT, 1 μl RNasin inhibitor, 4 μl NTP mixture (1 μl A,G,C, and 1 ul H2O), 100 μM UTP, 2 μl template (0.3 μg/μl), a-32P-UTP (10 μci/μl) and 1 μl SP6 polymerase (20 u/μl). Reaction time was 1 hour at 32° C. Unincorporated label and short nucleotides (<350base) were seperated from radiolabeled species by centrigugation on Chromaspin-200 columns (Clontech, Palo Alto, Calif.).
  • Cleavage reactions: synthetic RNA substrate was end-labeled with 32P using T4 polynucleotide kinase. Cleavage reaction system included 60 mM Tris-HCl (pH 7.5), 10 mM MgCl[0142] 2, 150 mM NaCl, 0.5 μM 32p-labled RNA oligo., 0.05-5 μM DNA enzyme. For cleavage of in vitro transcripts, reaction system contained 1% of PAI-1 transcripts, 25 mM Tris-HCl (pH 7.5), 5 mM MgCl2, 100 mM NaCl and 0.2-20 μM DNA enzyme. Reactions were allowed to proceed at 37° C. and were “quenched ” by transfer of aliquots to tubes containing 90% formamide, 20 mM EDTA and loading dye. Samples were separated by electrophoresis on TBE-urea denaturing polyacrylamide gels (5% gel for in vitro transcripts and 15% for synthetic RNA) and detected by autoradiography at −80° C.
  • DNA enzyme stability in serum: DAN enzymes were radiolabled using T4 polynucleotide kinase. Labeling reaction (20 μl of volume) consists of 1 [0143] μl 20 μM DNA enzyme, 2 μl 10× kinase buffer, 1 μl T4 PNK (10 u/μl), 10 μl H2O, and 6 μl 32P-ATP (3000 uCi/mmol, 10 uCi/μl). Reaction time is 30 minutes at 37° C. HUVEC were cultured in media (Clonetic) containing 2% or 20% FCS, and radiolabeled oligomers were added at a final concentration of 100 nM. Aliquots of the mixture were removed at different time points of 0, 3, 6, 12, 24 hours, quenched with phenol/chloroform and frozen until use. At the end of each experiment, all samples were phenol-extracted and analyzed by 15% denaturing polyacrylamide gels and visualized by autoradiography.
  • Culture conditions and DNA enzyme transfection: Primary HUVEC endothelial cells were obtained from Clonetic (USA) and grown in medium containing 2%FCS, 100 ug/ml streptomycin and 100 IU/ml penicillin at 37° C. in a humidified atmosphere of 5% CO2. HUVEC were used in experiments between [0144] passage 6 and 8. For transfection of DNA enzyme, rat endothelial cells (EC) were harvested and seeded into each well of 6-well plates (˜1×105 cells/well). Subconfluent (70-80%) EC were washed twice with 1 ml HEPES buffer (pH 7.4) and transfected using 0.5 ml of serum-free medium containing 1 μM test molecule (E2 or E0) and 20 μg/ml cationic lipids (DOTAP). 3 hours after incubation, 0.5 ml of 2% serum medium were added to each well; 3 hours later, TGFb was added to half the wells at a final concentration of 1.8 ng/ml. Transfected cells continued to be incubated for 8 hrs and were lysed to isolate RNA (for RT-PCR) and Protein (for Western blot) using Trizol reagent (Life Technologies).
  • RT-PCR: Total RNA from each well of 6-well plates were isolated using 1 ml Trizol reagent and dissolved in 20 μl Depc-treated H[0145] 2O. 2 μl samples were used to perform reverse transcription in 20 μl reaction system, and 4 μl products were then used as templates to amplify PAI-1 or human GAPDH in 50 μl PCR system (5 μl 10× buffer, 1 μl dNTP, 0.25 μl Taq polymerase, 1 μl forward and reverse primer, 38 μl H2O, 2 μl 32pdCTP (3000 ci/mmol, 10 uci/ul). The reaction conditions were 95° C.-2 min (predenatured), 95° C.-0.5 min, 58° C.-1 min, 72° C.-2 min, 35 cycles, 72° C.-10 min. Human GAPDH was used as internal control (forward Primer 5′TGAAGGTCGGAGTCAACGGATTTG3′; SEQ ID NO:13, reverse primer 5′CATGTGGGCCATGAGGTCCACCAC3′; SEQ ID NO:14), its PCR products being 452 base.
  • Western blot: Control and DNA enzyme-treated cells were harvested, washed with PBS and then the cell pellets were resuspended in lysis buffer and processed. Equal amounts of protein (15 μg/lane) were separated by SDS gel electrophoresis with 10% polyacrylamine separating gel using minigels (Bio-Rad). After electrophoresis, proteins were transferred to nitrocellulose membrane (Protran, Schleicler & Schuell) by electrotransfer and blocked for at least one hour at room temperature with 5% (W/V) BSA in TBS-T buffer (0.1M Tris-base (pH 7.5), 0.15M Nacl and 0.1% Tween-20) Following this step, membranes were immunoblotted with goat IgG polyclonal anti-PAI antibodies (Santa Cruz biotech) and then visualised by the ECL-system (Amershan) using horseradish peroxidase conjugated anti-goat IgG (Sigma). [0146]
  • Measurement of plasmin activity (PAI-1 functional assay): Conditioned medium and cell lysates were collected at various times after interventions. The cells were washed twice with PBS and lysed in 400 ml of 0.5% Triton X-100 for assaying plasmin. Conditioned media and Triton cell lysates were incubated with plasmin substrate Val-Leu-Lys-pNA (S2251, Kabivitru, Stockholm, Sweden) (final concentration, 0.35 mM), in a 96-well microtiter plate (final volume, 200 μl) for 60 min at room temperature. Plasmin activity was measured by reading the absorbance at 410 nm and was calculated against a plasmin (Sigma) standard regression line. [0147]
  • Animals, surgical procedures, injection of human cells, and quantitation of cellular migration into tissues: Rowett (rnu/rnu) athymic nude rats (Harlan Sprague Dawley, Indianapolis, Ind.) were used in studies approved by the “Columbia University Institute for Animal Care and Use Committee”. After anesthesia, a left thoracotomy was performed, the pericardium was opened, and the left anterior descending (LAD) coronary artery was ligated. At the time of surgery one group of rats received three intracardiac injections of E2 DNA enzyme, another received three intracardiac injections of E0 scrambled control, and a third group received three intracardiac injections of saline. For studies on neovascularization, 2.0×10[0148] 6 human cells obtained from a single donor after G-CSF mobilization were reconstituted with 2.0×105 immunopurified CD34+CD117bright cells, and injected into the rat tail vein 48 hours after LAD ligation. Each group consisted of 6-10 rats.
  • Quantitation of capillary density: In order to quantitate capillary density and species origin of the capillaries, additional sections were stained freshly with mAbs directed against factor VIII, rat or human CD31 (all Serotec, UK), and rat or human MHC class I (Accurate Chemicals, CT). Staining was performed by immunoperoxidase technique using an Avidin/Biotin Blocking Kit, a rat-absorbed biotinylated anti-mouse IgG, and a peroxidase-conjugate (all Vector Laboratories Burlingame, Calif.). Capillary density was determined from sections labeled with anti-Factor VIII mAb at 2 weeks post infarction and compared to the capillary density of the unimpaired myocardium. Values are expressed as factor VIII positive cells per HPF (600×). [0149]
  • Measurement of myocyte apoptosis by DNA end-labeling of paraffin tissue sections: For in situ detection of apoptosis at the single cell level we used the TUNEL method of DNA end-labeling mediated by dexynucleotidyl transferase (TdT) (Boehringer Mannheim, Mannheim, Germany). Rat myocardial tissue sections were obtained from LAD-ligated rats at two weeks after injection of either saline or CD34+ human cells, and from healthy rats as negative controls. Briefly, tissues were deparaffinized with xylene and rehydrated with graded dilutions of ethanol and two washes in phosphate-buffered saline (PBS). The tissue sections were then digested with Proteinase K (10 μg/ml in Tris/HCL) for 30 minutes at 37° C. The slides were then washed 3 times in PBS and incubated with 50 μl of the TUNEL reaction mixture (TdT and fluorescein-labeled dUTP) and incubated in a humid atmosphere for 60 minutes at 37° C. For negative controls TdT was eliminated from the reaction mixture. Following 3 washes in PBS, the sections were then incubated for 30 minutes with an antibody specific for fluorescein-conjugated alkaline phosphatase (AP) (Boehringer Mannheim, Mannheim, Germany). The TUNEL stain was visualized with a substrate system in which nuclei with DNA fragmentation stained blue, (BCIP/NBT substrate system, DAKO, Carpinteria, Calif.). The reaction was terminated following three minutes of exposure with PBS. To determine the proportion of blue-staining apoptotic nuclei within myocytes, tissue was counterstained with a monoclonal antibody specific for desmin. Endogenous peroxidase was blocked by using a 3% hydrogen perioxidase solution in PBS for 15 minutes, followed by washing with 20% goat serum solution. An anti-desmin antibody (Sigma, Saint Louis, Mo.) was incubated overnight (1:100) at 40° C. Following 3 washes sections were then treated with an anti-rabbit IgG, followed by a biotin conjugated secondary antibody for 30 minutes (Sigma, Saint Louis, Mo.). An avidin-biotin complex (Vector Laboratories, Burlingame, Calif.) was then added for an additional 30 minutes and the myocytes were visualized brown following 5 minutes exposure in DAB solution mixture (Sigma, Saint Louis, Mo.). Tissue sections were examined microscopically at 40× magnification and at least 100 cells were counted in a minimum of 8 high-power fields. The percentage of apoptotic myocytes was determined by means of an apoptotic index; the apoptotic index was calculated by dividing the number of positive staining myocyte nuclei by the total number of myocyte nuclei and multiplying by 100. Stained cells at the edges of the tissue were not counted. An apoptotic index of 1 or less was considered to indicate the absence of apoptosis. [0150]
  • Analyses of myocardial function: Echocardiographic studies were performed using a high frequency liner array transducer (SONOS 5500, Hewlett Packard, Andover, Mass.). 2D images were obtained at mid-papillary and apical levels. End-diastolic (EDV) and end-systolic (ESV) left ventricular volumes were obtained by bi-plane area-length method, and % left ventricular ejection fraction was calculated as [(EDV-ESV)/EDV]×100. Cardiac output (CO) was measured using an ultrasonic flowprobe (Transonic Systems Inc., Ithaca, N.Y.) and cardiac index was calculated as CO per weight. All echocardiographic studies were performed by a blinded investigator (ST). [0151]
  • REFERENCES
  • 1. Heymans, S. et al. Inhibition of plasminogen activators or matrix metalloproteinases prevents cardiac rupture but impairs therapeutic angiogenesis and causes cardiac failure. Nat. Med. 5, 1135-1142 (1999). [0152]
  • 2. Johnsen, M, Lund, L. R., Romer, J., Almholt, K, and Dano, K. Curr. Opin. Cell. Biol. 10, 667-671 (1998). [0153]
  • 3. Lijnen, H. R. et al. Arterioscler. Thromb. Vasc. Biol. 18, 1035-1045 (1998). [0154]
  • 4. Steffanson, S, and Lawrence, D. A. Nature 383, 441-443 (1996). [0155]
  • 5. Deng, G., royle, G., Wang, S., Crain, K., and Loskutoff, D. J. J. Biol. Chem. 272, 7678-7680 (1996). [0156]
  • 6. Stefansson, S. et al. J. Biol. Chem. 273, 6358-6366 (1998). [0157]
  • 7. Stefansson, S. et al. J. Biol. Chem. 276, 8135-8141 (2001). [0158]
  • 8. Brooks, P. C. et al. Cell 79, 1157-1164 (1994). [0159]
  • 9. Brooks, P. C., Clark, R. A. F., and Cheresh, D. A. Science 264,569-571 (1994). [0160]
  • 10. Bennett, M. R., and Schwartz, S. M. Antisense therapy for angioplasty restenosis: some critical considerations. Circulation 92,1981-1993 (1995). [0161]
  • 11. Stein, C. A. Is irrelevant cleavage the price of antisense efficacy? Pharmacology and Therapeutics 85, 231-236 (2000). [0162]
  • 12. Simayama, T., Nishikawa, F., Nishikawa, S., and Taira, K. Nuclease-resistant chimeric ribozymes containing deoxyribonucleotides snd phosphorothioate linkages. Nucleic Acid Res. 21, 2605-2611 (1993). [0163]
  • 13. Santoro, S. W., and Joyce, G. F. A general purpose RNA-cleaving DNA enzyme. Proc. Natl. Acad. Sci. USA. 94, 4262-4266 (1977). [0164]
  • 14. Santiago, F. S., et al. New DNA enzyme targeting Egr-1 mRNA inhibits vascular smooth muscle proliferation and regrowth after injury. [0165] Nat Med 5, 1264-1269 (1999).
  • 15. Breaker, R. R. Making catalytic DNAs. Science 290, 2095-2096 (2000). [0166]
  • 16. Khachigian, L. M. Catalytic DNAs as potential therapeutic agents and sequence-specific molecular tools to dissect biological function. J Clin Invest 106, 1189-1195 (2000). [0167]
  • 17. Zuker, M. On finding all suboptimal foldings of an RNA molecule. Science 244, 48-52 (1989). [0168]
  • 18. Soonpaa M H, Field L J. Assessment of cardiomyocyte DNA synthesis in normal and injured adult mouse hearts. Am J Physiol 272, H220-6 (1997). [0169]
  • 19. Kellerman S, Moore J A, Zierhut W, Zimmer H G, Campbell J, Gerdes A M. Nuclear DNA content and nucleation patterns in rat cardiac myocytes from different models of cardiac hypertrophy. J [0170] Mol Cell Cardiol 24, 497-505 (1992).
  • 20. Kajstura J, Leri A, Finato N, di Loreto N, Beltramo Calif., Anversa P. Myocyte proliferation in end-stage cardiac failure in humans. Proc Natl Acad Sci USA 95, 8801-8805 (1998). [0171]
  • 21. Beltrami A P, et al. Evidence that human cardiac myocytes divide after myocardial infarction. N Engl J Med 344, 1750-7 (2001). [0172]
  • 22. Matzura O, Wennborg A (1996) RNAdraw: an integrated program for RNA secondary structure calculation and analysis under 32-bit Microsoft Windows. [0173] Comput Appl Biosci 12:247-9.
  • 23. Santoro S W, Joyce G F (1997) A general purpose RNA-cleaving DNA enzyme. [0174] Proc Natl Acad Sci USA 94:4262-6.
  • 24. Santoro S W, Joyce G F (1998) Mechanism and utility of an RNA-cleaving DNA enzyme. [0175] Biochemistry 37:13330-42.
  • 25. Rossi et al., 1992, Aids Research and [0176] Human Retroviruses 8, 183.
  • 26. Hampel et al., EP0360257. [0177]
  • 27. Hampel and Tritz, 1989 Biochemistry 28, 4929. [0178]
  • 28. Hampel et al., 1990 Nucleic Acids Res. 18, 299. [0179]
  • 29. Perrotta and Been, 1992 [0180] Biochemistry 31, 16.
  • 30. Guerrier-Takada et al., 1983 Cell 35, 849. [0181]
  • 31. Forster and Altman, 1990 Science 249, 783. [0182]
  • 32. Saville and Collins, 1990 [0183] Cell 61, 685-696.
  • 33. Saville and Collins, 1991 Proc. Natl. Acad. Sci. USA 88, 8826-8830. [0184]
  • 34. Guo and Collins, 1995 EMBO J. 14, 368. [0185]
  • 35. Cech et al., U.S. Pat. No. 4,987,071. [0186]
  • 36. M., Andreasen, P. A., Nielsen, L., Dano, K., Lebo, R. V. Gelehrter, T. D. (1986) “cDNA cloning of human plasminogen activator-inhibitor from endothelial cells” J. Clin. Invest. 78(6):1673-1680. [0187]
  • 37. Zhang et al. (1995) Clinical Pharmacology & Therapeutics 58(1), 44-53. [0188]
  • 38. Sambrook et al., “Molecular Cloning: A Laboratory Manual”, Second Edition (1989), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. [0189]
  • 39. Ma L, Fogo A B. (2001) “Role of angiotensin II in glomerular injury” Semin. Nephrol. 21(6):544-53. [0190]
  • 40. Rerolle J P, Hertig A, Nguyen G, Sraer J D, Rondeau E P. (2000) Kidney Int. 58(5):1841-50. [0191]
  • 41. Epstein M. (2001) “Aldosterone as a mediator of progressive renal dysfunction: evolving perspectives”. Intern. Med. 40(7):573-83. [0192]
  • 42. Oda T, Jung Y O, Kim H S, Cai X, Lopez-Guisa J M, Ikeda Y, Eddy A A. (2001) “PAI-1 deficiency attenuates the fibrogenic response to ureteral obstruction.” Kidney Int. 60(2):587-96. [0193]
  • 43. Tahashi Y, Matsuzaki K, Date M, Yoshida K, Furukawa F, Sugano Y, Matsushita M, Himeno Y, Inagaki Y, Inoue K. (2002) “Differential regulation of TGF-beta signal in hepatic stellate cells between acute and chronic rat liver injury.” Hepatology 35(1):49-61. [0194]
  • 44. Zhang L P, Takahara T, Yata Y, Furui K, Jin B, Kawada N, Watanabe A. (1999) J. Hepatol. 31(4):703-11. [0195]
  • 45. Kotani I, Sato A, Hayakawa H, Urano T, Takada Y, Takada (1999) A. Thromb. Res. 77(6):493-504. [0196]
  • [0197]
  • 1 17 1 15 DNA ARTIFICIAL SEQUENCE DNAzyme Catalytic Domain 1 ggctagctac aacga 15 2 31 DNA ARTIFICIAL SEQUENCE DNAzyme for human Plasminogen Activator Inhibitor-1 2 catctgcagg ctagctacaa cgacctgaag t 31 3 33 DNA ARTIFICIAL SEQUENCE DNAzyme for human Plasminogen Activator Inhibitor-1 3 ctggagacag gctagctaca acgactgcat cct 33 4 31 DNA ARTIFICIAL SEQUENCE DNAzyme for Plasminogen Activator Inhibitor 4 gctgaagagg ctagctacaa cgaatctgca t 31 5 2876 DNA Homo Sapiens 5 gaattcctgc agctcagcag ccgccgccag agcaggacga accgccaatc gcaaggcacc 60 tctgagaact tcaggatgca gatgtctcca gccctcacct gcctagtcct gggcctggcc 120 cttgtctttg gtgaagggtc tgctgtgcac catcccccat cctacgtggc ccacctggcc 180 tcagacttcg gggtgagggt gtttcagcag gtggcgcagg cctccaagga ccgcaacgtg 240 gttttctcac cctatggggt ggcctcggtg ttggccatgc tccagctgac aacaggagga 300 gaaacccagc agcagattca agcagctatg ggattcaaga ttgatgacaa gggcatggcc 360 cccgccctcc ggcatctgta caaggagctc atggggccat ggaacaagga tgagatcagc 420 accacagacg cgatcttcgt ccagcgggat ctgaagctgg tccagggctt catgccccac 480 ttcttcaggc tgttccggag cacggtcaag caagtggact tttcagaggt ggagagagcc 540 agattcatca tcaatgactg ggtgaagaca cacacaaaag gtatgatcag caacttgctt 600 gggaaaggag ccgtggacca gctgacacgg ctggtgctgg tgaatgccct ctacttcaac 660 ggccagtgga agactccctt ccccgactcc agcacccacc gccgcctctt ccacaaatca 720 gacggcagca ctgtctctgt gcccatgatg gctcagacca acaagttcaa ctatactgag 780 ttcaccacgc ccgatggcca ttactacgac atcctggaac tgccctacca cggggacacc 840 ctcagcatgt tcattgctgc cccttatgaa aaagaggtgc ctctctctgc cctcaccaac 900 attctgagtg cccagctcat cagccactgg aaaggcaaca tgaccaggct gccccgcctc 960 ctggttctgc ccaagttctc cctggagact gaagtcgacc tcaggaagcc cctagagaac 1020 ctgggaatga ccgacatgtt cagacagttt caggctgact tcacgagtct ttcagaccaa 1080 gagcctctcc acgtcgcgca ggcgctgcag aaagtgaaga tcgaggtgaa cgagagtggc 1140 acggtggcct cctcatccac agctgtcata gtctcagccc gcatggcccc cgaggagatc 1200 atcatggaca gacccttcct ctttgtggtc cggcacaacc ccacaggaac agtccttttc 1260 atgggccaag tgatggaacc ctgaccctgg ggaaagacgc cttcatctgg gacaaaactg 1320 gagatgcatc gggaaagaag aaactccgaa gaaaagaatt ttagtgttaa tgactctttc 1380 tgaaggaaga gaagacattt gccttttgtt aaaagatggt aaaccagatc tgtctccaag 1440 accttggcct ctccttggag gacctttagg tcaaactccc tagtctccac ctgagaccct 1500 gggagagaag tttgaagcac aactccctta aggtctccaa accagacggt gacgcctgcg 1560 ggaccatctg gggcacctgc ttccacccgt ctctctgccc actcgggtct gcagacctgg 1620 ttcccactga ggccctttgc aggatggaac tacggggctt acaggagctt ttgtgtgcct 1680 ggtagaaact atttctgttc cagtcacatt gccatcactc ttgtactgcc tgccaccgcg 1740 gaggaggctg gtgacaggcc aaaggccagt ggaagaaaca ccctttcatc tcagagtcca 1800 ctgtggcact ggccacccct ccccagtaca ggggtgctgc aggtggcaga gtgaatgtcc 1860 cccatcatgt ggcccaactc tcctggcctg gccatctccc tccccagaaa cagtgtgcat 1920 gggttatttt ggagtgtagg tgacttgttt actcattgaa gcagatttct gcttcctttt 1980 atttttatag gaatagagga agaaatgtca gatgcgtgcc cagctcttca ccccccaatc 2040 tcttggtggg gaggggtgta cctaaatatt tatcatatcc ttgcccttga gtgcttgtta 2100 gagagaaaga gaactactaa ggaaaataat attatttaaa ctcgctccta gtgtttcttt 2160 gtggtctgtg tcaccgtatc tcaggaagtc cagccacttg actggcacac acccctccgg 2220 acatccagcg tgacggagcc cacactgcca ccttgtggcc gcctgagacc ctcgcgcccc 2280 ccgcgccccc cgcgcccctc tttttcccct tgatggaaat tgaccataca atttcatcct 2340 ccttcagggg atcaaaagga cggagtgggg ggacagagac tcagatgagg acagagtggt 2400 ttccaatgtg ttcaatagat ttaggagcag aaatgcaagg ggctgcatga cctaccagga 2460 cagaactttc cccaattaca gggtgactca cagccgcatt ggtgactcac ttcaatgtgt 2520 catttccggc tgctgtgtgt gagcagtgga cacgtgaggg gggggtgggt gagagagaca 2580 ggcagctcgg attcaactac cttagataat atttctgaaa acctaccagc cagagggtag 2640 ggcacaaaga tggatgtaat gcactttggg aggccaaggc gggaggattg cttgagccca 2700 ggagttcaag accagcctgg gcaacatacc aagacccccg tctctttaaa aatatatata 2760 ttttaaatat acttaaatat atatttctaa tatctttaaa tatatatata tattttaaag 2820 accaatttat gggagaattg cacacagatg tgaaatgaat gtaatctaat agaagc 2876 6 402 PRT HOMO SAPIENS 6 Met Gln Met Ser Pro Ala Leu Thr Cys Leu Val Leu Gly Leu Ala Leu 1 5 10 15 Val Phe Gly Glu Gly Ser Ala Val His His Pro Pro Ser Tyr Val Ala 20 25 30 His Leu Ala Ser Asp Phe Gly Val Arg Val Phe Gln Gln Val Ala Gln 35 40 45 Ala Ser Lys Asp Arg Asn Val Val Phe Ser Pro Tyr Gly Val Ala Ser 50 55 60 Val Leu Ala Met Leu Gln Leu Thr Thr Gly Gly Glu Thr Gln Gln Gln 65 70 75 80 Ile Gln Ala Ala Met Gly Phe Lys Ile Asp Asp Lys Gly Met Ala Pro 85 90 95 Ala Leu Arg His Leu Tyr Lys Glu Leu Met Gly Pro Trp Asn Lys Asp 100 105 110 Glu Ile Ser Thr Thr Asp Ala Ile Phe Val Gln Arg Asp Leu Lys Leu 115 120 125 Val Gln Gly Phe Met Pro His Phe Phe Arg Leu Phe Arg Ser Thr Val 130 135 140 Lys Gln Val Asp Phe Ser Glu Val Glu Arg Ala Arg Phe Ile Ile Asn 145 150 155 160 Asp Trp Val Lys Thr His Thr Lys Gly Met Ile Ser Asn Leu Leu Gly 165 170 175 Lys Gly Ala Val Asp Gln Leu Thr Arg Leu Val Leu Val Asn Ala Leu 180 185 190 Tyr Phe Asn Gly Gln Trp Lys Thr Pro Phe Pro Asp Ser Ser Thr His 195 200 205 Arg Arg Leu Phe His Lys Ser Asp Gly Ser Thr Val Ser Val Pro Met 210 215 220 Met Ala Gln Thr Asn Lys Phe Asn Tyr Thr Glu Phe Thr Thr Pro Asp 225 230 235 240 Gly His Tyr Tyr Asp Ile Leu Glu Leu Pro Tyr His Gly Asp Thr Leu 245 250 255 Ser Met Phe Ile Ala Ala Pro Tyr Glu Lys Glu Val Pro Leu Ser Ala 260 265 270 Leu Thr Asn Ile Leu Ser Ala Gln Leu Ile Ser His Trp Lys Gly Asn 275 280 285 Met Thr Arg Leu Pro Arg Leu Leu Val Leu Pro Lys Phe Ser Leu Glu 290 295 300 Thr Glu Val Asp Leu Arg Lys Pro Leu Glu Asn Leu Gly Met Thr Asp 305 310 315 320 Met Phe Arg Gln Phe Gln Ala Asp Phe Thr Ser Leu Ser Asp Gln Glu 325 330 335 Pro Leu His Val Ala Gln Ala Leu Gln Lys Val Lys Ile Glu Val Asn 340 345 350 Glu Ser Gly Thr Val Ala Ser Ser Ser Thr Ala Val Ile Val Ser Ala 355 360 365 Arg Met Ala Pro Glu Glu Ile Ile Met Asp Arg Pro Phe Leu Phe Val 370 375 380 Val Arg His Asn Pro Thr Gly Thr Val Leu Phe Met Gly Gln Val Met 385 390 395 400 Glu Pro 7 30 DNA ARTIFICIAL SEQUENCE SCRAMBLED CONTROL SEQUENCE 7 acactggagg ctagcacaac gatgacgagt 30 8 21 RNA Homo Sapiens 8 aacuucagga ugcagauguc u 21 9 21 RNA Homo Sapiens 9 aggaugcaga ugucuccagc c 21 10 24 RNA Rattus Sp. 10 gaugcagaug ucuucagccc ucau 24 11 22 DNA ARTIFICIAL SEQUENCE PRIMER derived from human source 11 ccaagagcgc tgtcaagaag ac 22 12 22 DNA ARTIFICIAL SEQUENCE PRIMER derived from human source 12 tcaccgtctg ctttggagac ct 22 13 24 DNA ARTIFICIAL SEQUENCE PRIMER derived from human source 13 tgaaggtcgg agtcaacgga tttg 24 14 24 DNA ARTIFICIAL SEQUENCE PRIMER derived from human source 14 catgtgggcc atgaggtcca ccac 24 15 3053 DNA Rattus Norvegicus 15 cccccgagag ctttgtgaag gaggaacgct gcacacccgc ctcccgcagc acacagccaa 60 ccacagctga gcgacacgca acaagagcca atcacaaggc acttccgaaa gctccaggat 120 gcagatgtct tcagccctca cttgcctcac cctgggcctg gttctggtct ttgggaaagg 180 gttcgcttca ccccttccag agtcccatac agcccagcag gccaccaact tcggagtaaa 240 agtgtttcag catgtggtcc aggcctccaa agaccgaaat gtggtcttct ctccctacgg 300 cgtgtcctcg gtgctggcta tgctgcagct gaccacagca gggaaaaccc ggcagcagat 360 ccaagatgct atgggattca atatcagtga gaggggcaca gctcctgccc tccgaaagct 420 ctccaaggag ctcatggggt catggaacaa gaatgagatc agtactgcgg acgccatctt 480 tgtccagcgg gacctagagc tggtccaggg cttcatgccc cacttcttca agctcttccg 540 gaccacggtg aagcaggtgg acttctcaga ggtggaaaga gccagattca tcatcaacga 600 ctgggtggag aggcacacca aaggtatgat cagtgactta ctggccaagg gggctgtaaa 660 tgagctgaca cgcctggtgc tggtgaacgc cctctatttc aacggccaat ggaagacccc 720 cttcttagag gccagcaccc accagcgcct gttccacaag tctgatggta gcaccatctc 780 cgtgcccatg atggctcaga acaacaagtt caactacact gagttcacca ctccggatgg 840 gcacgagtac gacatcctgg aactgcccta ccacggcgaa accctcagca tgttcattgc 900 agcacccttt gaaaaagatg tgcccctctc cgccatcacc aacattttgg acgctgagct 960 catcagacaa tggaagagca acatgaccag gctgccccgc ctcctcatcc tgcctaagtt 1020 ctctctggag actgaagtgg acctcagagg gcccctggag aagctgggca tgactgacat 1080 cttcagctca acccaggccg acttcacaag tctttccgac caagagcagc tctctgtagc 1140 acaagcacta caaaaggtca agatcgaggt gaacgagagc ggcacagtgg cgtcttcctc 1200 cacagccatt ctagtctcag cccgcatggc ccccacggag atggttttag accgatcctt 1260 tctctttgtg gttcggcaca atccaacaga gacaatcctc ttcatgggcc agctgatgga 1320 gccttgagag tgggatgaga agcctttcct ttgggacaaa actggacgtg ttataagcag 1380 agactctgaa gaaaagaatt gttttaagga ctctttgggg agaaagagaa ggcctttctt 1440 tcttaccccg gcactggtaa atctttccaa ccagcctccc agacctcaga ctctcgaaga 1500 ggaaagagtc taactccctc actagggacc tatcttacta aggtctcatc caaccataga 1560 actcacagaa tctggatctg cccagcattc agcctttgga cccagttccc accaaggccc 1620 cagcagggcc aacccactac gccttcactc agcaaagtct tttgtgttcc agtcacactc 1680 tgggtacctc ttgtatcgtc ctccattgct atgaaggatg acccaggcca aaggaagaag 1740 cactgtccta tctcaaggtc cactgtggaa atgaacacct tgcccatccc caaggggcag 1800 cagatagaca gatcgaatga tcgcccgata tcaagccttc tcccagctcc cgtcctgccc 1860 tcccttccct gacagccgcc ttgtgttatt tcagagtgta gatgacttgt ttacagcttt 1920 tttcgaccca caaacttttc tcattttgaa agcgtgaaag aaaggtcaga tgtgcacgtg 1980 ccttgctctt tatcctgggt ctccctgtga ggggagaggg gtcctgggga gattccaggg 2040 gtgtgattga atatttatct tgtttatctt atacgtttgt tggggagaag aagcactatt 2100 aaggagaaag ccttttattt aaaccatggc atatggtgtc ccatttgggg tctgtatccc 2160 tgtatgtcag ggaggcatca ctccacaaac ccgcccctcg ggtggcccgg cgtcggggct 2220 cacactgccg cctagtggca gccgaacacg cccttgcccc atccctcccc cgcatcctcc 2280 cccgtggctc ttttccttag ggatcttgcc aaggtgatgc ttggcagccc acggtaaagg 2340 aagggggaaa aagattaggt gggagagaga gagagagaga gagagagaga gagagagaga 2400 gagagagaga gagagagaga gagagagaga aagagagaga gatgtttgag agagggcaaa 2460 gtggtttcaa atttttccaa tacattcaga agccgagtgg gaaagggggc tgtgtgacct 2520 aacaggacag aactttctcc aattactggg tgactcagct gcactggtga ctcacttcaa 2580 tgtgtcattt ccggctgctg taagtgagca gtggacacgt gggggggggg gggtgaggat 2640 gaaagaaaca gccagctcct ggtcaaccac cttagttaga taatcttttt tgaaagcttc 2700 ctagctgggg gtatgatcag aaaaccaatt tactgaaaaa ctgcacagga aggtaacgtg 2760 aatctaattt catagcgggc cgctctgcat ccgttacatc tccactggaa aaaaataatc 2820 attttctttt tgtgtgtgtg tgtgtgtttt agcttttctc cctctccctc tttctctctc 2880 atttcattat gcactggata accatacacc gtgtaccaca ggggcccaaa tgtggggtca 2940 catggtcttg aattttgtgg ggtacatatg cctttgtttg tttgttttca cttttgatat 3000 ataaacaggt aaatgtgttt ttaaaaaata ataaaaatag agaatatgca gac 3053 16 402 PRT Rattus Norvegicus 16 Met Gln Met Ser Ser Ala Leu Thr Cys Leu Thr Leu Gly Leu Val Leu 1 5 10 15 Val Phe Gly Lys Gly Phe Ala Ser Pro Leu Pro Glu Ser His Thr Ala 20 25 30 Gln Gln Ala Thr Asn Phe Gly Val Lys Val Phe Gln His Val Val Gln 35 40 45 Ala Ser Lys Asp Arg Asn Val Val Phe Ser Pro Tyr Gly Val Ser Ser 50 55 60 Val Leu Ala Met Leu Gln Leu Thr Thr Ala Gly Lys Thr Arg Gln Gln 65 70 75 80 Ile Gln Asp Ala Met Gly Phe Asn Ile Ser Glu Arg Gly Thr Ala Pro 85 90 95 Ala Leu Arg Lys Leu Ser Lys Glu Leu Met Gly Ser Trp Asn Lys Asn 100 105 110 Glu Ile Ser Thr Ala Asp Ala Ile Phe Val Gln Arg Asp Leu Glu Leu 115 120 125 Val Gln Gly Phe Met Pro His Phe Phe Lys Leu Phe Arg Thr Thr Val 130 135 140 Lys Gln Val Asp Phe Ser Glu Val Glu Arg Ala Arg Phe Ile Ile Asn 145 150 155 160 Asp Trp Val Glu Arg His Thr Lys Gly Met Ile Ser Asp Leu Leu Ala 165 170 175 Lys Gly Ala Val Asn Glu Leu Thr Arg Leu Val Leu Val Asn Ala Leu 180 185 190 Tyr Phe Asn Gly Gln Trp Lys Thr Pro Phe Leu Glu Ala Ser Thr His 195 200 205 Gln Arg Leu Phe His Lys Ser Asp Gly Ser Thr Ile Ser Val Pro Met 210 215 220 Met Ala Gln Asn Asn Lys Phe Asn Tyr Thr Glu Phe Thr Thr Pro Asp 225 230 235 240 Gly His Glu Tyr Asp Ile Leu Glu Leu Pro Tyr His Gly Glu Thr Leu 245 250 255 Ser Met Phe Ile Ala Ala Pro Phe Glu Lys Asp Val Pro Leu Ser Ala 260 265 270 Ile Thr Asn Ile Leu Asp Ala Glu Leu Ile Arg Gln Trp Lys Ser Asn 275 280 285 Met Thr Arg Leu Pro Arg Leu Leu Ile Leu Pro Lys Phe Ser Leu Glu 290 295 300 Thr Glu Val Asp Leu Arg Gly Pro Leu Glu Lys Leu Gly Met Thr Asp 305 310 315 320 Ile Phe Ser Ser Thr Gln Ala Asp Phe Thr Ser Leu Ser Asp Gln Glu 325 330 335 Gln Leu Ser Val Ala Gln Ala Leu Gln Lys Val Lys Ile Glu Val Asn 340 345 350 Glu Ser Gly Thr Val Ala Ser Ser Ser Thr Ala Ile Leu Val Ser Ala 355 360 365 Arg Met Ala Pro Thr Glu Met Val Leu Asp Arg Ser Phe Leu Phe Val 370 375 380 Val Arg His Asn Pro Thr Glu Thr Ile Leu Phe Met Gly Gln Leu Met 385 390 395 400 Glu Pro 17 1209 DNA HOMO SAPIENS 17 atgcagatgt ctccagccct cacctgccta gtcctgggcc tggcccttgt ctttggtgaa 60 gggtctgctg tgcaccatcc cccatcctac gtggcccacc tggcctcaga cttcggggtg 120 agggtgtttc agcaggtggc gcaggcctcc aaggaccgca acgtggtttt ctcaccctat 180 ggggtggcct cggtgttggc catgctccag ctgacaacag gaggagaaac ccagcagcag 240 attcaagcag ctatgggatt caagattgat gacaagggca tggcccccgc cctccggcat 300 ctgtacaagg agctcatggg gccatggaac aaggatgaga tcagcaccac agacgcgatc 360 ttcgtccagc gggatctgaa gctggtccag ggcttcatgc cccacttctt caggctgttc 420 cggagcacgg tcaagcaagt ggacttttca gaggtggaga gagccagatt catcatcaat 480 gactgggtga agacacacac aaaaggtatg atcagcaact tgcttgggaa aggagccgtg 540 gaccagctga cacggctggt gctggtgaat gccctctact tcaacggcca gtggaagact 600 cccttccccg actccagcac ccaccgccgc ctcttccaca aatcagacgg cagcactgtc 660 tctgtgccca tgatggctca gaccaacaag ttcaactata ctgagttcac cacgcccgat 720 ggccattact acgacatcct ggaactgccc taccacgggg acaccctcag catgttcatt 780 gctgcccctt atgaaaaaga ggtgcctctc tctgccctca ccaacattct gagtgcccag 840 ctcatcagcc actggaaagg caacatgacc aggctgcccc gcctcctggt tctgcccaag 900 ttctccctgg agactgaagt cgacctcagg aagcccctag agaacctggg aatgaccgac 960 atgttcagac agtttcaggc tgacttcacg agtctttcag accaagagcc tctccacgtc 1020 gcgcaggcgc tgcagaaagt gaagatcgag gtgaacgaga gtggcacggt ggcctcctca 1080 tccacagctg tcatagtctc agcccgcatg gcccccgagg agatcatcat ggacagaccc 1140 ttcctctttg tggtccggca caaccccaca ggaacagtcc ttttcatggg ccaagtgatg 1200 gaaccctga 1209

Claims (38)

What is claimed is:
1. A catalytic nucleic acid that specifically cleaves an mRNA encoding a Plasminogen Activator Inhibitor-1 (PAI-1) comprising, in 5′ to 3′ order:
(a) consecutive nucleotides defining a first binding domain of at least 4 nucleotides;
(b) consecutive nucleotides defining a catalytic domain located contiguous with the 3′ end of the first binding domain, and capable of cleaving the PAI-1-encoding mRNA at a predetermined phosphodiester bond; and
(c) consecutive nucleotides defining a second binding domain of at least 4 nucleotides located contiguous with the 3′ end of the catalytic domain,
wherein the sequence of the nucleotides in each binding domain is complementary to a sequence of ribonucleotides in the PAI-1-encoding mRNA and wherein the catalytic nucleic acid hybridizes to and specifically cleaves the PAI-1-encoding mRNA.
2. The catalytic nucleic acid of claim 1, wherein the nucleotides of the first binding domain comprise at least one deoxyribonucleotide.
3. The catalytic nucleic acid of claim 1, wherein the nucleotides of the second binding domain comprise at least one deoxyribonucleotide.
4. The catalytic nucleic acid of claim 1, wherein the nucleotides of the first binding domain comprise at least one deoxyribonucleotide derivative.
5. The catalytic nucleic acid of claim 1, wherein the nucleotides of the second binding domain comprise at least one deoxyribonucleotide derivative.
6. The catalytic nucleic acid of claim 1, wherein the nucleotides of the first binding domain comprise at least one ribonucleotide.
7. The catalytic nucleic acid of claim 1, wherein the nucleotides of the second binding domain comprise at least one ribonucleotide.
8. The catalytic nucleic acid of claim 1, wherein the nucleotides of the first binding domain comprise at least one ribonucleotide derivative.
9. The catalytic nucleic acid of claim 1, wherein the nucleotides of the second binding domain comprise at least one ribonucleotide derivative.
10. The catalytic nucleic acid of claim 1, wherein the nucleotides of the first binding domain comprise at least one modified base.
11. The catalytic nucleic acid of claim 1, wherein the nucleotides of the second binding domain comprise at least one modified base.
12. The catalytic nucleic acid of claim 1, wherein the nucleotides of the first binding domain comprise at least one modified internucleoside bond.
13. The catalytic nucleic acid of claim 1, wherein the nucleotides of the second binding domain comprise at least one modified internucleoside bond.
14. The catalytic nucleic acid of claim 12 or 13, wherein the modified internucleoside bond is a phosphorothioate bond.
15. The catalytic nucleic acid of claim 1, wherein the PAI-1-encoding mRNA encodes human PAI-1.
16. The catalytic nucleic acid of claim 15, wherein the human PAI-1-encoding mRNA has the sequence set forth in SEQ ID NO:5.
17. A pharmaceutical composition comprising the catalytic nucleic acid of claim 1 and a pharmaceutically acceptable carrier.
18. A method of specifically inhibiting the expression of PAI-1 in a cell that would otherwise express PAI-1, comprising contacting the cell with the catalytic nucleic acid of claim 1 so as to specifically inhibit the expression of PAI-1 in the cell.
19. A method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the catalytic nucleic acid of claim 1 effective to specifically inhibit the expression of PAI-1 in the subject's cells.
20. A method of specifically inhibiting the expression of PAI-1 in a subject's cells comprising administering to the subject an amount of the pharmaceutical composition of claim 17 effective to specifically inhibit the expression of PAI-1 in the subject's cells.
21. A method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the pharmaceutical composition of claim 17 effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease.
22. A method of treating a fibrotic disease in a subject involving fibrogenesis which comprises administering to the subject an amount of the pharmaceutical composition of claim 17 effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease.
23. The method of claim 22, wherein the fibrotic disease is a renal disease.
24. The method of claim 22, wherein the fibrotic disease is a hepatic disease.
25. The method of claim 22, wherein the fibrotic disease is a disease of the lung.
26. The method of claim 22, wherein the fibrotic disease is a disease of the skin.
27. The method of claim 22, wherein the fibrotic disease is a disease of the eye.
28. An oligonucleotide comprising consecutive nucleotides that hybridizes with a PAI-1-encoding mRNA under conditions of high stringency and is between 8 and 40 nucleotides in length.
29. The oligonucleotide of claim 28, wherein at least one internucleoside linkage within the oligonucleotide comprises a phosphorothioate linkage.
30. The oligonucleotide of claim 28, wherein the nucleotides comprise at least one deoxyribonucleotide.
31. The oligonucleotide of claim 28, wherein the nucleotides comprise at least one ribonucleotide.
32. The oligonucleotide of claim 28, wherein the PAI-1-encoding mRNA encodes human PAI-1.
33. The oligonucleotide of claim 32, wherein the human PAI-1-encoding mRNA comprises consecutive nucleotides, the sequence of which is set forth in SEQ ID NO:5.
34. A method of treating a subject which comprises administering to the subject an amount of the oligonucleotide of claim 28 effective to inhibit expression of a PAI-1 in the subject so as to thereby treat the subject.
35. A method of treating a cardiovascular disease in a subject involving apoptosis of a cardiomyocyte in the subject which comprises administering to the subject an amount of the oligonucleotide of claim 28 effective to inhibit apoptosis of the cardiomyocyte in the subject so as to thereby treat the cardiovascular disease.
36. A method of treating a fibrotic disease in a subject involving fibrogenesis in the subject which comprises administering to the subject an amount of the oligonucleotide of claim 28 effective to inhibit fibrogenesis in the subject so as to thereby treat the fibrotic disease.
37. The method of any of claim 19, 20, 21, 22, 34, 35, or 36, wherein the subject is a mammal.
38. The method of claim 37, wherein the mammal is a human being.
US10/128,706 2002-04-23 2002-04-23 DNA enzyme to inhibit plasminogen activator inhibitor-1 Abandoned US20030199463A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
US10/128,706 US20030199463A1 (en) 2002-04-23 2002-04-23 DNA enzyme to inhibit plasminogen activator inhibitor-1
ZA200408776A ZA200408776B (en) 2002-04-23 2003-04-23 A DNA enzyme to inhibit plasminogen activator inhibitor-1
AU2003231089A AU2003231089B2 (en) 2002-04-23 2003-04-23 A DNA enzyme to inhibit plasminogen activator inhibitor-1
PCT/US2003/012767 WO2003091456A1 (en) 2002-04-23 2003-04-23 A dna enzyme to inhibit plasminogen activator inhibitor-1
CN038147149A CN1662663B (en) 2002-04-23 2003-04-23 DNA enzyme to inhibit plasminogen activator inhibitor-1
US10/512,496 US7662794B2 (en) 2002-04-23 2003-04-23 DNA enzyme to inhibit plasminogen activator inhibitor-1
AT03724216T ATE477324T1 (en) 2002-04-23 2003-04-23 DNA ENZYME FOR INHIBITING PLASMINOGEN ACTIVATOR INHIBITOR-1
JP2003587981A JP4574992B2 (en) 2002-04-23 2003-04-23 DNA enzyme for inhibiting plasminogen activator inhibitor-1
DE60333747T DE60333747D1 (en) 2002-04-23 2003-04-23 DNA ENZYME FOR INHIBITING PLASMINOGENACTIVATOR INHIBITOR-1
CA2483007A CA2483007C (en) 2002-04-23 2003-04-23 A dna enzyme to inhibit plasminogen activator inhibitor-1
EP03724216A EP1501948B1 (en) 2002-04-23 2003-04-23 A dna enzyme to inhibit plasminogen activator inhibitor-1
IL164674A IL164674A (en) 2002-04-23 2004-10-18 Catalytic deoxyribonucleic acid that cleaves pai-1 and uses thereof
AU2009201027A AU2009201027B2 (en) 2002-04-23 2009-03-04 A DNA enzyme to inhibit plasminogen activator inhibitor-1

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10/128,706 US20030199463A1 (en) 2002-04-23 2002-04-23 DNA enzyme to inhibit plasminogen activator inhibitor-1

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10512496 Continuation-In-Part 2003-04-23

Publications (1)

Publication Number Publication Date
US20030199463A1 true US20030199463A1 (en) 2003-10-23

Family

ID=29215497

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/128,706 Abandoned US20030199463A1 (en) 2002-04-23 2002-04-23 DNA enzyme to inhibit plasminogen activator inhibitor-1
US10/512,496 Expired - Lifetime US7662794B2 (en) 2002-04-23 2003-04-23 DNA enzyme to inhibit plasminogen activator inhibitor-1

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/512,496 Expired - Lifetime US7662794B2 (en) 2002-04-23 2003-04-23 DNA enzyme to inhibit plasminogen activator inhibitor-1

Country Status (11)

Country Link
US (2) US20030199463A1 (en)
EP (1) EP1501948B1 (en)
JP (1) JP4574992B2 (en)
CN (1) CN1662663B (en)
AT (1) ATE477324T1 (en)
AU (2) AU2003231089B2 (en)
CA (1) CA2483007C (en)
DE (1) DE60333747D1 (en)
IL (1) IL164674A (en)
WO (1) WO2003091456A1 (en)
ZA (1) ZA200408776B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050148527A1 (en) * 2002-04-23 2005-07-07 Silviu Itescu Dna enzyme to inhibit plasminogen activator inhibitor-1

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2791144C (en) * 2006-07-05 2019-04-09 Catalyst Biosciences, Inc. Protease screening methods and proteases identified thereby
KR20170014023A (en) 2006-07-05 2017-02-07 카탈리스트 바이오사이언시즈, 인코포레이티드 Protease screening methods and proteases identified thereby
CA2711878A1 (en) * 2008-01-09 2009-07-16 Intrexon Corporation Therapeutic inhibitors of pai-1 function and methods of their use
CN101549150A (en) * 2008-03-31 2009-10-07 刘建宁 Application of prourokinase and prourokinase mutant in acute myocardial infarction facilitated percutaneous coronary artery intervention

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5985558A (en) * 1997-04-14 1999-11-16 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the inibition of c-Jun and c-Fos

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU5720498A (en) 1996-12-24 1998-07-17 Ribozyme Pharmaceuticals, Inc. Chemical synthesis of nucleosides analogs and their incorporation into polynucleotides
AU2001229441A1 (en) 2000-01-14 2001-07-24 Tanox, Inc. Use of antagonists of plasminogen activator inhibitor-1 (pai-1) for the treatment of asthma and chronic obstructive pulmonary disease
AU3173301A (en) * 2000-02-11 2001-08-20 European Molecular Biology Laboratory Methods and compositions for treatment of alzheimer's disease by enhancing plasmin or plasmin-like activity
US20030199463A1 (en) * 2002-04-23 2003-10-23 Silviu Itescu DNA enzyme to inhibit plasminogen activator inhibitor-1

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5985558A (en) * 1997-04-14 1999-11-16 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the inibition of c-Jun and c-Fos

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050148527A1 (en) * 2002-04-23 2005-07-07 Silviu Itescu Dna enzyme to inhibit plasminogen activator inhibitor-1
US7662794B2 (en) 2002-04-23 2010-02-16 The Trustees Of Columbia University In The City Of New York DNA enzyme to inhibit plasminogen activator inhibitor-1

Also Published As

Publication number Publication date
AU2009201027B2 (en) 2012-02-23
CA2483007C (en) 2018-05-22
WO2003091456A1 (en) 2003-11-06
EP1501948A4 (en) 2007-05-23
JP2005523708A (en) 2005-08-11
AU2009201027A1 (en) 2009-04-02
CA2483007A1 (en) 2003-11-06
US20050148527A1 (en) 2005-07-07
JP4574992B2 (en) 2010-11-04
DE60333747D1 (en) 2010-09-23
CN1662663B (en) 2012-09-26
US7662794B2 (en) 2010-02-16
ZA200408776B (en) 2007-12-27
CN1662663A (en) 2005-08-31
IL164674A (en) 2012-01-31
AU2003231089B2 (en) 2008-12-04
IL164674A0 (en) 2005-12-18
ATE477324T1 (en) 2010-08-15
EP1501948A1 (en) 2005-02-02
AU2003231089A1 (en) 2003-11-10
EP1501948B1 (en) 2010-08-11

Similar Documents

Publication Publication Date Title
US6566127B1 (en) Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor
US6127173A (en) Nucleic acid catalysts with endonuclease activity
US6831171B2 (en) Nucleic acid catalysts with endonuclease activity
US7235534B2 (en) Antisense strategy to modulate estrogen receptor response (ER α and/or ER β )
US7034009B2 (en) Enzymatic nucleic acid-mediated treatment of ocular diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R)
JP3054745B2 (en) Antisense oligonucleotide regulation of raf gene expression
JPH10500309A (en) Methods and compositions for treating restenosis and cancer using ribozymes
AU2021204399A1 (en) Compositions and methods for inhibiting gene expression of alpha-1 antitrypsin
US20190169618A1 (en) Nucleic acids and methods for the treatment of pompe disease
US6037463A (en) Enzymatic RNA molecules that cleave mutant N-RAS
AU2009201027B2 (en) A DNA enzyme to inhibit plasminogen activator inhibitor-1
JP2003525037A (en) Methods and reagents for regulation and diagnosis of CD20 and NOGO gene expression
AU749561B2 (en) Nucleic acid molecules having endonuclease and/or catalytic activity
US20040072783A1 (en) Nucleozymes with endonuclease activity
JP2003521943A (en) Nucleozymes having endonuclease activity
US6656731B1 (en) Nucleic acid catalysts with endonuclease activity
Prins et al. Antisense of oligonucleotides and the inhibition of oncogene expression
US20030087847A1 (en) Method and reagent for the inhibition of checkpoint kinase-1 (Chk1) enzyme
US20100324116A1 (en) Fas/fasl or other death receptor targeted methods and compositions for killing tumor cells
US7078390B1 (en) Ribozymes to growth factor originating in human platelet
AU3986201A (en) Method and reagent for the inhibition of grid
EP1321521A1 (en) Ribozymes directed against flt-1

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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