US20060094677A1 - Inhibition of apoptosis-specific eIF-5A ("eIF-5A1") with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics - Google Patents

Inhibition of apoptosis-specific eIF-5A ("eIF-5A1") with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics Download PDF

Info

Publication number
US20060094677A1
US20060094677A1 US11/134,445 US13444505A US2006094677A1 US 20060094677 A1 US20060094677 A1 US 20060094677A1 US 13444505 A US13444505 A US 13444505A US 2006094677 A1 US2006094677 A1 US 2006094677A1
Authority
US
United States
Prior art keywords
apoptosis
cell
cells
specific eif
sirna
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
US11/134,445
Other versions
US20100168047A9 (en
Inventor
John Thompson
Bruce Galton
Catherine Taylor
Charles Dinarello
Leonid Reznikov
Adrienne Boone
Marianne Hopkins
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.)
Eloxx Pharmaceuticals Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/909,796 external-priority patent/US6867237B1/en
Priority claimed from US10/141,647 external-priority patent/US7166467B2/en
Priority claimed from US10/200,148 external-priority patent/US7968523B2/en
Priority claimed from US10/277,969 external-priority patent/US7217517B2/en
Priority claimed from US10/383,614 external-priority patent/US7381708B2/en
Priority to US11/134,445 priority Critical patent/US20100168047A9/en
Application filed by Individual filed Critical Individual
Priority to US11/184,982 priority patent/US20060154887A1/en
Assigned to SENESCO TECHNOLOGIES, INC. reassignment SENESCO TECHNOLOGIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REZNIKOV, LEONID, GALTON, BRUCE C., BOONE, ADRIENNE, DINARELLO, CHARLES, HOPKINS, MARIANNE, TAYLOR, CATHERINE, THOMPSON, JOHN E.
Publication of US20060094677A1 publication Critical patent/US20060094677A1/en
Assigned to YA GLOBAL INVESTMENTS, L.P. reassignment YA GLOBAL INVESTMENTS, L.P. SECURITY AGREEMENT Assignors: SENESCO TECHNOLOGIES, INC., SENESCO, INC.
Publication of US20100168047A9 publication Critical patent/US20100168047A9/en
Priority to US13/099,171 priority patent/US8754057B2/en
Priority to US14/475,389 priority patent/US20140371299A1/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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • 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
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • A61P9/10Drugs 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
    • 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
    • C12N2310/11Antisense
    • 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
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed

Definitions

  • the present invention relates to apoptosis-specific eucaryotic initiation factor (“eIF-5A”) or referred to as “apoptosis-specific eIF-5A” or “eIF-5A1” and deoxyhypusine synthase (DHS).
  • eIF-5A eucaryotic initiation factor
  • DHS deoxyhypusine synthase
  • the present invention relates to apoptosis-specific eIF-5A and DHS nucleic acids and polypeptides and methods for inhibiting expression of apoptosis-specific eIF-5A and DHS.
  • Apoptosis is a genetically programmed cellular event that is characterized by well-defined morphological features, such as cell shrinkage, chromatin condensation, nuclear fragmentation, and membrane blebbing. Kerr et al. (1972) Br. J. Cancer, 26, 239-257; Wyllie et al. (1980) Int. Rev. Cytol., 68, 251-306. It plays an important role in normal tissue development and homeostasis, and defects in the apoptotic program are thought to contribute to a wide range of human disorders ranging from neurodegenerative and autoimmunity disorders to neoplasms. Thompson (1995) Science, 267, 1456-1462; Mullauer et al. (2001) Mutat. Res, 488, 211-231. Although the morphological characteristics of apoptotic cells are well characterized, the molecular pathways that regulate this process have only begun to be elucidated.
  • caspases cysteine proteases
  • Creagh & Martin 2001) Biochem. Soc. Trans, 29, 696-701; Dales et al. (2001) Leuk. Lymphoma, 41, 247-253.
  • Caspases trigger apoptosis in response to apoptotic stimuli by cleaving various cellular proteins, which results in classic manifestations of apoptosis, including cell shrinkage, membrane blebbing and DNA fragmentation. Chang & Yang (2000) Microbiol. Mol. Biol. Rev., 64, 821-846.
  • Pro-apoptotic proteins such as Bax or Bak
  • caspase-activating molecules such as mitochondrial cytochrome c
  • Anti-apoptotic proteins such as Bcl-2, promote cell survival by antagonizing the activity of the pro-apoptotic proteins, Bax and Bak. Tsujimoto (1998) Genes Cells, 3, 697-707; Kroemer (1997) Nature Med., 3, 614-620.
  • the ratio of Bax:Bcl-2 is thought to be one way in which cell fate is determined; an excess of Bax promotes apoptosis and an excess of Bcl-2 promotes cell survival.
  • Another key protein involved in apoptosis is a protein that encoded by the tumor suppressor gene p53. This protein is a transcription factor that regulates cell growth and induces apoptosis in cells that are damaged and genetically unstable, presumably through up-regulation of Bax.
  • Bold et al. (1997) Surgical Oncology, 6, 133-142; Ronen et al., 1996; Schuler & Green (2001) Biochem. Soc. Trans., 29, 684-688; Ryan et al. (2001) Curr. Opin. Cell Biol., 13, 332-337; Zörmig et al. (2001) Biochem. Biophys. Acta, 1551, F1-F37.
  • Cytokines also have been implicated in the apoptotic pathway. Biological systems require cellular interactions for their regulation, and cross-talk between cells generally involves a large variety of cytokines. Cytokines are mediators that are produced in response to a wide variety of stimuli by many different cell types. Cytokines are pleiotropic molecules that can exert many different effects on many different cell types, but are especially important in regulation of the immune response and hematopoietic cell proliferation and differentiation. The actions of cytokines on target cells can promote cell survival, proliferation, activation, differentiation, or apoptosis depending on the particular cytokine, relative concentration, and presence of other mediators.
  • anti-cytokines to treat autoimmune disorders such as psoriasis, rheumatoid arthritis, and Crohn's disease is gaining popularity.
  • Anti-cytokine therapies that reduce the biological activities of these two cytokines can provide therapeutic benefits (Dinarello and Abraham, 2002).
  • Interleukin 1 is an important cytokine that mediates local and systemic inflammatory reactions and which can synergize with TNF in the pathogenesis of many disorders, including vasculitis, osteoporosis, neurodegenerative disorders, diabetes, lupus nephritis, and autoimmune disorders such as rheumatoid arthritis.
  • IL-1 ⁇ Interleukin 1
  • the importance of IL-1 ⁇ in tumour angiogenesis and invasiveness was also recently demonstrated by the resistance of IL-1 ⁇ knockout mice to metastases and angiogenesis when injected with melanoma cells (Voronov et al., 2003).
  • Interleukin 18 is a recently discovered member of the IL-1 family and is related by structure, receptors, and function to IL-1.
  • IL-18 is a central cytokine involved in inflammatory and autoimmune disorders as a result of its ability to induce interferon-gamma (IFN- ⁇ ), TNF- ⁇ , and IL-1.
  • IFN- ⁇ interferon-gamma
  • TNF- ⁇ TNF- ⁇
  • IL-1 ⁇ and IL-18 are both capable of inducing production of TNF- ⁇ , a cytokine known to contribute to cardiac dysfunction during myocardial ischemia (Maekawa et al., 2002).
  • Inhibition of IL-18 by neutralization with an IL-18 binding protein was found to reduce ischemia-induced myocardial dysfunction in an ischemia/reperfusion model of suprafused human atrial myocardium (Dinarello, 2001).
  • Neutralization of IL-18 using a mouse IL-18 binding protein was also able to decrease IFN- ⁇ , TNF- ⁇ , and IL-1 ⁇ transcript levels and reduce joint damage in a collagen-induced arthritis mouse model (Banda et al., 2003).
  • a reduction of IL-18 production or availability may also prove beneficial to control metastatic cancer as injection of IL-18 binding protein in a mouse melanoma model successfully inhibited metastases (Carrascal et al., 2003).
  • IL-18 As a further indication of its importance as a pro-inflammatory cytokine, plasma levels of IL-18 were elevated in patients with chronic liver disease and increased levels were correlated with the severity of the disease (Ludwiczek et aL., 2002). Similarly, IL-18 and TNF- ⁇ were elevated in the serum of diabetes mellitus patients with nephropathy (Moriwaki et al., 2003). Neuroinflammation following traumatic brain injury is also mediated by pro-inflammatory cytokines and inhibition of IL-18 by the IL-18 binding protein improved neurological recovery in mice following brain trauma (Yatsiv et al., 2002).
  • TNF- ⁇ a member of the TNF family of cytokines, is a pro-inflammatory cytokine with pleiotropic effects ranging from co-mitogenic effects on hematopoietic cells, induction of inflammatory responses, and induction of cell death in many cell types.
  • TNF- ⁇ is normally induced by bacterial lipopolysaccharides, parasites, viruses, malignant cells and cytokines and usually acts beneficially to protect cells from infection and cancer.
  • inappropriate induction of TNF- ⁇ is a major contributor to disorders resulting from acute and chronic inflammation such as autoimmune disorders and can also contribute to cancer, AIDS, heart disease, and sepsis (reviewed by Aggarwal and Natarajan, 1996; Sharma and Anker, 2002).
  • TNF- ⁇ TNF- ⁇
  • B-CLL B-cell chronic lymphocytic leukemia
  • IL-1 ⁇ Interleukin-1 ⁇
  • DHS Deoxyhypusine synthase
  • eIF-5A hypusine-containing eucaryotic translation initiation Factor-5A
  • Hypusine a unique amino acid, is found in all examined eucaryotes and archaebacteria, but not in eubacteria, and eIF-5A is the only known hypusine-containing protein.
  • Active eIF-5A is formed in two post-translational steps: the first step is the formation of a deoxyhypusine residue by the transfer of the 4-aminobutyl moiety of spermidine to the ⁇ -amino group of a specific lysine of the precursor eIF-5A catalyzed by deoxyhypusine synthase; the second step involves the hydroxylation of this 4-aminobutyl moiety by deoxyhypusine hydroxylase to form hypusine.
  • eIF-5A intracellular depletion results in a significant accumulation of specific mRNAs in the nucleus, indicating that eIF-5A may be responsible for shuttling specific classes of mRNAs from the nucleus to the cytoplasm.
  • the accumulation of eIF-5A at nuclear pore-associated intranuclear filaments and its interaction with a general nuclear export receptor further suggest that eIF-5A is a nucleocytoplasmic shuttle protein, rather than a component of polysomes. Rosorius et al. (1999) J. Cell Science, 112, 2369-2380.
  • the first cDNA for eIF-5A was cloned from human in 1989 by Smit-McBride et al., and since then cDNAs or genes for eIF-5A have been cloned from various eukaryotes including yeast, rat, chick embryo, alfalfa, and tomato.
  • eIF-5A mRNA has been explored in various human tissues and mammalian cell lines. For example, changes in eIF-5A expression have been observed in human fibroblast cells after addition of serum following serum deprivation. Pang & Chen (1994) J. Cell Physiol., 160, 531-538. Age-related decreases in deoxyhypusine synthase activity and abundance of precursor eIF-5A have also been observed in senescing fibroblast cells, although the possibility that this reflects averaging of differential changes in isoforms was not determined. Chen & Chen (1997) J. Cell Physiol., 170, 248-254.
  • eIF-5A may be the cellular target of viral proteins such as the human immunodeficiency virus type 1 Rev protein and human T cell leukemia virus type 1 Rex protein. Ruhl et al. (1993) J. Cell Biol.,123, 1309-1320; Katahira et al. (1995) J. Virol., 69, 3125-3133. Preliminary studies indicate that eIF-5A may target RNA by interacting with other RNA-binding proteins such as Rev, suggesting that these viral proteins may recruit eIF-5A for viral RNA processing. Liu et al. (1997) Biol. Signals, 6, 166-174.
  • eIF5A and DHS are known, there remains a need in understanding how these proteins are involved in apoptotic pathways as well as cytokine stimulation to be able to modulate apoptosis and cytokine expression.
  • the present invention fulfills this need.
  • the present invention relates to apoptosis specific eucaryotic initiation factor 5A (eIF-5A), referred to as “apoptosis specific eIF-5A” or “eIF-5A1.”
  • eIF-5A eucaryotic initiation factor 5A
  • the present invention also relates to apoptosis-specific eIF-5A nucleic acids and polypeptides and methods for inhibiting or suppressing apoptosis in cells using antisense nucleotides or siRNAs to inhibit expression of apoptosis-specific eIF-5A.
  • the present invention relates to a method of delivering siRNA to mammalian lung cells in vivo.
  • the invention also relates to suppressing or inhibiting expression of pro-inflammatory cytokines by inhibiting expression of apoptosis-specific eIF-5A. Further, the present invention relates to inhibiting or suppressing expression of p53 by inhibiting expression of apoptosis-specific eIF-5A. The present invention also relates to a method of increasing Bcl-2 expression by inhibiting or suppression expression of apoptosis factor 5A using antisense nucleotides or siRNAs. The present invention also provides a method of inhibiting production of cytokines, especially TNF- ⁇ in human epithelial cells.
  • suppressing expression of apoptosis-specific eIF-5A by the use of antisense oligonucleotides targeted at apoptosis-specific eIF-5A provides methods of preventing retinal ganglion cell death in a glaucomatous eye.
  • FIG. 1 depicts the nucleotide sequence (SEQ ID NO: 11) and derived amino acid sequence (SEQ ID NO: 12) of the 3′ end of rat apoptosis-specific eIF-5A.
  • FIG. 2 depicts the nucleotide sequence (SEQ ID NO: 15) and derived amino acid sequence (SEQ ID NO: 16) of the 5′ end of rat apoptosis-specific eIF-5A cDNA.
  • FIG. 3 depicts the nucleotide sequence of rat corpus luteum apoptosis-specific eIF-5A full-length cDNA (SEQ ID NO: 1). The amino acid sequence is shown in SEQ ID NO: 2.
  • FIG. 4 depicts the nucleotide sequence (SEQ ID NO: 6) and derived amino acid sequence (SEQ ID NO: 7) of the 3′ end of rat apoptosis-specific DHS cDNA.
  • FIG. 5 is an alignment of the full-length nucleotide sequence of rat corpus luteum apoptosis-specific eIF-5A cDNA (SEQ ID NO: 20) with the nucleotide sequence of human eIF-5A (SEQ ID NO: 3) (Accession number BC000751 or NM — 001970, SEQ ID NO:3).
  • FIG. 6 is an alignment of the full-length nucleotide sequence of rat corpus luteum apoptosis-specific eIF-5A cDNA (SEQ ID NO: 20) with the nucleotide sequence of human eIF-5A (SEQ ID NO: 4) (Accession number NM-020390, SEQ ID NO:4).
  • FIG. 7 is an alignment of the full-length nucleotide sequence of rat corpus luteum apoptosis-specific eIF-5A cDNA (SEQ ID NO: 20) with the nucleotide sequence of mouse eIF-5A (Accession number BC003889).
  • Mouse nucleotide sequence (Accession number BC003889) is SEQ ID NO:5.
  • FIG. 8 is an alignment of the derived full-length amino acid sequence of rat corpus luteum apoptosis-specific eIF-5A (SEQ ID NO: 2) with the derived amino acid sequence of human eIF-5A (SEQ ID NO: 21) (Accession number BC000751 or NM — 001970).
  • FIG. 9 is an alignment of the derived full-length amino acid sequence of rat corpus luteum apoptosis-specific eIF-5A (SEQ ID NO: 2) with the derived amino acid sequence of human eIF-5A (SEQ ID NO: 22) (Accession number NM — 020390).
  • FIG. 10 is an alignment of the derived full-length amino acid sequence of rat corpus luteum apoptosis-specific eIF-5A (SEQ ID NO: 2) with the derived amino acid sequence of mouse eIF-5A (SEQ ID NO: 23) (Accession number BC003889).
  • FIG. 11 is an alignment of the partial-length nucleotide sequence of rat corpus luteum apoptosis-specific DHS cDNA (residues 1-453 of SEQ ID NO: 6) with the nucleotide sequence of human DHS (SEQ ID NO: 8) (Accession number BC000333, SEQ ID NO:8).
  • FIG. 12 is a restriction map of rat corpus luteum apoptosis-specific eIF-5A cDNA.
  • FIG. 13 is a restriction map of the partial-length rat apoptosis-specific DHS cDNA.
  • FIG. 14 is a Northern blot (top) and an ethidium bromide stained gel (bottom) of total RNA probed with the 32 P-dCTP-labeled 3′-end of rat corpus luteum apoptosis-specific eIF-5A cDNA.
  • FIG. 15 is a Northern blot (top) and an ethidium bromide stained gel (bottom) of total RNA probed with the 32 P-dCTP-labeled 3′-end of rat corpus luteum apoptosis-specific DHS cDNA.
  • FIG. 16 depicts a DNA laddering experiment in which the degree of apoptosis in superovulated rat corpus lutea was examined after injection with PGF-2 ⁇ .
  • FIG. 17 is an agarose gel of genomic DNA isolated from apoptosing rat corpus luteum showing DNA laddering after treatment of rats with PGF F-2 ⁇ .
  • FIG. 18 depicts a DNA laddering experiment in which the degree of apoptosis in dispersed cells of superovulated rat corpora lutea was examined in rats treated with sperrnidine prior to exposure to PGF-2 ⁇ .
  • FIG. 19 depicts a DNA laddering experiment in which the degree of apoptosis in superovulated rat corpus lutea was examined in rats treated with spermidine and/or PGF-2 ⁇ .
  • FIG. 20 is a Southern blot of rat genomic DNA probed with 32 P-dCTP-labeled partial-length rat corpus luteum apoptosis-specific eIF-5A cDNA.
  • FIG. 21 depicts pHM6, a mammalian epitope tag expression vector (Roche Molecular Biochemicals).
  • FIG. 22 is a Northern blot (top) and ethidium bromide stained gel (bottom) of total RNA isolated from COS-7 cells after induction of apoptosis by withdrawal of serum probed with the 32 P-dCTP-labeled 3′-untranslated region of rat corpus luteum apoptosis-specific DHS cDNA.
  • FIG. 23 is a flow chart illustrating the procedure for transient transfection of COS-7 cells.
  • FIG. 24 is a Western blot of transient expression of foreign proteins in COS-7 cells following transfection with pHM6.
  • FIG. 25 illustrates enhanced apoptosis as reflected by increased caspase activity when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 26 illustrates enhanced apoptosis as reflected by increased DNA fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 27 illustrates detection of apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 28 illustrates enhanced apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 29 illustrates detection of apoptosis as reflected by phosphatidylserine exposure when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 30 illustrates enhanced apoptosis as reflected by increased phosphatidylserine exposure when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 31 illustrates enhanced apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 32 illustrates enhanced apoptosis when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 33 illustrates down-regulation of Bcl-2 when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • the top photo is the Coomassie-blue-stained protein blot; the bottom photo is the corresponding Western blot.
  • FIG. 34 is a Coomassie-blue-stained protein blot and the corresponding Western blot of COS-7 cells transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the antisense orientation using Bcl-2 as a probe.
  • FIG. 35 is a Coomassie-blue-stained protein blot and the corresponding Western blot of COS-7 cells transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation using c-Myc as a probe.
  • FIG. 36 is a Coomassie-blue-stained protein blot and the corresponding Western blot of COS-7 cells transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation when p53 is used as a probe.
  • FIG. 37 is a Coomassie-blue-stained protein blot and the corresponding Western blot of expression of pHM6-full-length rat apoptosis-specific eIF-5A in COS-7 cells using an anti-[HA]-peroxidase probe and a Coomassie-blue-stained protein blot and the corresponding Western blot of expression of pHM6-full-length rat apoptosis-specific eIF-5A in COS-7 cells when a p53 probe is used.
  • FIG. 38 is an alignment of human eIF5A2 isolated from RKO cells (SEQ ID NO: 24) with the sequence of human eIF5A2 (SEQ ID NO: 22) (Genbank accession number XM — 113401). The consensus sequence is shown in SEQ ID NO: 28.
  • FIG. 39 is a graph depicting the percentage of apoptosis occurring in RKO and RKO-E6 cells following transient transfection.
  • RKO and RKO-E6 cells were transiently transfected with pHM6-LacZ or pHM6-apoptosis-specific eIF-5A.
  • RKO cells treated with Actinomycin D and transfected with pHM6-apoptosis-specific eIF-5A showed a 240% increase in apoptosis relative to cells transfected with pHM6-LacZ that were not treated with Actinomycin D.
  • FIG. 40 is a graph depicting the percentage of apoptosis occurring in RKO cells following transient transfection.
  • RKO cells were transiently transfected with pHM6-LacZ, pHM6-apoptosis-specific eIF-5A, pHM6-eIF5A2, or pHM6-truncated apoptosis-specific eIF-5A.
  • Cells transfected with pHM6-apoptosis-specific eIF-5A showed a 25% increase in apoptosis relative to control cells transfected with pHM6-LacZ. This increase was not apparent for cells transfected with pHM6-eIF5A2 or pHM6-truncated apoptosis-specific eIF-5A.
  • FIG. 41 is a graph depicting the percentage of apoptosis occurring in RKO cells following transient transfection.
  • RKO cells were either left untransfected or were transiently transfected with pHM6-LacZ or pHM6-apoptosis-specific eIF-5A. After correction for transfection efficiency, 60% of the cells transfected with pHM6-apoptosis-specific eIF-5A were apoptotic.
  • FIG. 42 provides the results of a flow cytometry analysis of RKO cell apoptosis following transient transfection.
  • RKO cells were either left untransfected or were transiently transfected with pHM6-LacZ, pHM6-apoptosis-specific eIF-5A, pHM6-eIF5A2, or pHM6-truncated apoptosis-specific eIF-5A.
  • the table depicts the percentage of cells undergoing apoptosis calculated based on the area under the peak of each gate. After correction for background apoptosis in untransfected cells and for transfection efficiency, 80% of cells transfected with pHM6-apoptosis-specific eIF-5A exhibited apoptosis.
  • Cells transfected with pHM6-LacZ, pHM6-eIF5A2 or pHM6-truncated apoptosis-specific eIF-5A exhibited only background levels of apoptosis.
  • FIG. 43 provides Western blots of protein extracted from RKO cells treated with 0.25 ⁇ g/ml Actinomycin D for 0, 3, 7, 24, and 48 hours.
  • the top panel depicts a Western blot using anti-p53 as the primary antibody.
  • the middle panel depicts a Western blot using anti-apoptosis-specific eIF-5A as the primary antibody.
  • the bottom panel depicts the membrane used for the anti-apoptosis-specific eIF-5A blot stained with Coomassie blue following chemiluminescent detection to demonstrate equal loading.
  • p53 and apoptosis-specific eIF-5A are both upregulated by treatment with Actinomycin D.
  • FIG. 44 is a bar graph showing that both apoptosis-specific eIF-5A and proliferation eIF-5A are expressed in heart tissue.
  • the heart tissue was taken from patients receiving coronary artery bypass grafts (“CABG”).
  • CABG coronary artery bypass grafts
  • Gene expression levels apoptosis-specific eIF-5A (light gray bar) are compared to proliferation eIF-5A (dark gray bar).
  • the X-axis is patient identifier numbers.
  • the Y-axis is pg/ng of 18s (picograms of message RNA over nanograms of ribosomal RNA 18S).
  • FIG. 45 is a bar graph showing that both apoptosis-specific eIF-5A and proliferation eIF-5A are expressed in heart tissue.
  • the heart tissue was taken from patients receiving valve replacements.
  • Gene expression levels of apoptosis-specific eIF-5A (light gray bar) are compared to proliferation eIF-5A (dark gray bar).
  • the X-axis is patient identifier numbers.
  • the Y-axis is pg/ng of 18s (picograms of message RNA over nanograms of ribosomal RNA 18S).
  • FIG. 46 is a bar graph showing the gene expression levels measured by real-time PCR of apoptosis-specific eIF-5A (elf5a) versus proliferation eIF-5A (eIF5b) in pre-ischemia heart tissue and post ischemia heart tissue.
  • the Y-axis is pg/ng of 18s (picograms of message RNA over nanograms of ribosomal RNA 18S).
  • FIG. 47 depicts schematically an experiment performed on heart tissue.
  • the heart tissue was exposed to normal oxygen levels and the expression levels apoptosis-specific eIF-5A and proliferating eIF-5A measured. Later, the amount of oxygen delivered to the heart tissue was lowered, thus inducing hypoxia and ischemia, and ultimately, a heart attack in the heart tissue.
  • the expression levels of apoptosis-specific eIF-5A and proliferating eIF-5A were measured and compared to the expression levels of the heart tissue before it was damaged by ischemia.
  • FIG. 48 shows EKGs of heart tissue before and after the ischemia was induced.
  • FIG. 49 shows the lab bench with the set up of the experiment depicted in FIG. 47 .
  • FIGS. 50 A-F report patient data where the levels of apoptosis-specific eIF-5A are correlated with levels of IL-1 ⁇ and IL-18.
  • FIG. 50A is a chart of data obtained from coronary artery bypass graft (CABG) patients.
  • FIG. 50B is a chart of data obtained from valve replacement patients.
  • FIG. 50C is a graph depicting the correlation of apoptosis-specific eIF-5A to IL-18 in CABG patients.
  • FIG. 50D is a graph depicting the correlation of proliferating eIF-5A to IL-18 in CABG patients.
  • FIG. 50E is a graph depicting the correlation of apoptosis-specific eIF-5A to IL-18 in valve replacement patients.
  • FIG. 50F is a graph depicting the correlation of proliferating eIF-5A to IL-18 in valve replacement patients.
  • FIG. 51 is a chart of the patient's data from which patients data used in FIGS. 50 A-F was obtained.
  • FIG. 52 shows the levels of protein produced by RKO cells after being treated with antisense oligo 1, 2 and 3 (of apoptosis-specific eIF-5A)(SEQ ID NO: 35, 37 and 39, respectively).
  • the RKO cells produced less apoptosis-specific eIF-5A as well as less p53 after having been transfected with the antisense apoptosis-specific eIF-5A oligonucleotides.
  • FIG. 53 shows uptake of the fluorescently labeled antisense oligonucleotide.
  • FIGS. 54-58 show a decrease in the percentage of cells undergoing apoptosis in the cells having being treated with antisense apoptosis-specific eIF-5A oligonucleotides as compared to cells not having been transfected with the antisense apoptosis-specific eIF-5A oligonucleotides.
  • FIG. 59 shows that treating lamina cribrosa cells with TNF- ⁇ and/or camptothecin caused an increase in the number of cells undergoing apoptosis.
  • FIGS. 60 and 61 shows a decrease in the percentage of cells undergoing apoptosis in the cells having being treated with antisense apoptosis-specific eIF-5A oligonucleotides as compared to cells not having been transfected with the antisense apoptosis-specific eIF-5A oligonucleotides.
  • FIG. 62 shows that the lamina cribrosa cells uptake the labeled siRNA either in the presence of serum or without serum.
  • FIG. 63 shows that cells transfected with apoptosis-specific eIF-5A siRNA produced less apoptosis-specific eIF-5A protein and in addition, produced more Bcl-2 protein. A decrease in apoptosis-specific eIF-5A expression correlates with an increase in BCL-2 expression.
  • FIG. 64 shows that cells transfected with apoptosis-specific eIF-5A siRNA produced less apoptosis factor 5a protein.
  • FIGS. 65-67 shows that cells transfected with apoptosis-specific eIF-5A siRNA had a lower percentage of cells undergoing apoptosis after exposure to amptothecin and TNF- ⁇ than untransfected cells.
  • FIG. 68 are photographs of Hoescht-stained lamina cribrosa cell line #506 transfected with siRNA and treated with camptothecin and TNF- ⁇ from the experiment described in
  • FIG. 67 and Example 13 The apoptosing cells are seen as more brightly stained cells. They have smaller nucleic because of chromatin condensation and are smaller and irregular in shape.
  • FIG. 69 shows that IL-1 exposed HepG2 cells transfected with apoptosis-specific eIF-5A cells secreted less TNF- ⁇ than non-transfected cells.
  • FIG. 70 shows the sequence of human apoptosis-specific eIF-5A (SEQ ID NO:29) and the sequences of 5 siRNAs of the present invention (SEQ ID NO:30, 31, 32, 33 and 34).
  • FIG. 71 shows the sequence of human apoptosis-specific eIF-5A (SEQ ID NO: 29) and the sequences of 3 antisense oligonucleotides of the present invention (SEQ ID NO:35, 37, and 39, respectively in order of appearance ).
  • FIG. 72 shows the binding position of three antisense oligonucleotides (SEQ ID NO:25-27, respectively in order of appearance) targeted against human apoptosis-specific eIF-5A.
  • the full-length nucleotide sequence is SEQ ID NO: 19.
  • FIG. 73 a and b shows the nucleotide alignment (SEQ ID NO: 41 and 42, respectively in order of appearance) and amino acid alignment (SEQ ID NO: 43 and 22, respectively in order of appearance) of human apoptosis-specific eIF-5A against human proliferating eIF-5A.
  • FIG. 74A provides a picture of a Western blot where siRNAs against apoptosis-specific eIF-5A have reduced if not inhibited the production of TNF- ⁇ in transfected HT-29 cells.
  • FIG. 74B provides the results of an ELISA.
  • FIG. 75 provides the results of an ELISA. TNF- ⁇ production was reduced in cells treated with siRNAs against apoptosis-specific eIF-5A as compared to control cells.
  • FIG. 76 shows the time course of the U-937 differentiation experiment. See Example 16.
  • FIG. 77 shows the results of a Western blot showing that apoptosis-specific eIF-5A is up-regulated during monocyte differentiation and subsequence TNF- ⁇ secretion.
  • FIG. 78 depicts stem cell differentiation and the use of siRNAs against apoptosis-specific eIF-5A to inhibit cytokine production.
  • FIG. 79 is a bar graph showing that IL-8 is produced in response to TNF-alpha as well as in response to interferon. This graph shows that siRNA against apoptosis-specific eIF-5A blocked almost all IL-8 produced in response to interferon as well as a significant amount of the IL-8 produced as a result of the combined treatment of interferon and TNF.
  • FIG. 80 is another bar graph showing that IL-8 is produced in response to TNF-alpha as well as in response to interferon. This graph shows that siRNA against apoptosis-specific eIF-5A blocked almost all IL-8 produced in response to interferon as well as a significant amount of the IL-8 produced as a result of the combined treatment of interferon and TNF.
  • FIG. 81 is a western blot of HT-29 cells treated with IFN gamma for 8 and 24 hours. This blot shows up-regulation in HT-29 cells (4 fold at 8 hours) of against apoptosis-specific eIF-5A in response to interferon gamma.
  • FIG. 82 is a characterization of lamina cribrosa cells by immunofluorescence.
  • Lamina cribrosa cells (#506) isolated from the optic nerve head of an 83-year old male were characterized by immunofluorescence.
  • Primary antibodies were a) actin; b) fibronectin; c) laminin; d) GFAP. All pictures were taken at 400 times magnification.
  • FIG. 83 is a graph showing percent apoptosis of lamina cribrosa cell line #506 in response to treatment with camptothecin and TNF- ⁇ .
  • Lamina cribrosa cell line #506 cells were seeded at 40,000 cells per well onto an 8-well culture slide. Three days later the confluent LC cells were treated with either 10 ng/ml TNF- ⁇ , 50 ⁇ M camptothecin, or 10 ng/ml TNF- ⁇ plus 50 ⁇ M camptothecin. An equivalent volume of DMSO, a vehicle control for camptothecin, was added to the untreated control cells. The cells were stained with Hoescht 33258 48 hours after treatment and viewed by fluorescence microscopy using a UV filter. Cells with brightly stained condensed or fragmented nuclei were counted as apoptotic.
  • FIG. 84 shows expression levels of against apoptosis-specific eIF-5A during camptothecin or TNF-a plus camptothecin treatment.
  • Lamina cribrosa cell #506 cells were seeded at 40,000 cells per well onto a 24-well plate. Three days later the LC cells were treated with either 50 ⁇ M camptothecin or 10 ng/ml TNF- ⁇ plus 50 ⁇ M camptothecin and protein lysate was harvested 1, 4, 8, and 24 hours later. An equivalent volume of DMSO was added to control cells as a vehicle control and cell lysate was harvested 1 and 24 hours later.
  • FIG. 85 shows expression levels of apoptosis-specific eIF-5A in lamina cribosa cell lines #506 and #517 following transfection with siRNAs.
  • Lamina cribrosa cell #506 and #517 cells were seeded at 10,000 cells per well onto a 24-well plate.
  • the LC cells were transfected with either GAPDH siRNA, apoptosis-specific eIF-5A siRNAs #1-4 (SEQ ID NO:30-33) or control siRNA #5 (SEQ ID NO:34).
  • the protein lysate was harvested and 5 ⁇ g of protein from each sample was separated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with anti-eIF5A antibody.
  • FIG. 86 shows the percent apoptosis of lamina cribosa cell line #506 cells transfected with apoptosis-specific eIF-5A siRNAs and treated with TNF- ⁇ and camptothecin.
  • Lamina cribrosa cell line #506 cells were seeded at 7500 cells per well onto an 8-well culture slide. Three days later the LC cells were transfected with either GAPDH siRNA, apoptosis-specific eIF-5A siRNAs #1-4 (SEQ ID NO:30-33), or control siRNA #5 (SEQ ID NO:34). 72 hours after transfection, the transfected cells were treated with 10 ng/ml TNF- ⁇ plus 50 ⁇ M camptothecin.
  • FIG. 87 shows percent apoptosis of lamina cribosa cell line #517 cells transfected with apoptosis-specific eIF-5A siRNA #1 and treated with TNF- ⁇ and camptothecin.
  • Lamina cribrosa cell line #517 cells were seeded at 7500 cells per well onto an 8-well culture slide. Three days later the LC cells were transfected with either apoptosis-specific eIF-5A siRNA #1 (SEQ ID NO:30) or control siRNA #5 (SEQ ID NO:34). 72 hours after transfection, the transfected cells were treated with 10 ng/ml TNF- ⁇ plus 50 ⁇ M camptothecin.
  • FIG. 88 shows TUNEL-labeling of lamina cribosa cell line #506 cells transfected with apoptosis-specific eIF-5A siRNA #1 and treated with TNF- ⁇ and camptothecin.
  • Lamina cribrosa cell line #506 cells were seeded at 7500 cells per well onto an 8-well culture slide. Three days later the LC cells were transfected with either apoptosis-specific eIF-5A siRNA #1 (SEQ ID NO:30) or control siRNA #5 (SEQ ID NO:34). 72 hours after transfection, the transfected cells were treated with 10 ng/ml TNF- ⁇ plus 50 ⁇ M camptothecin.
  • TUNEL terminal deoxynucleotidyl transferase-mediated dUTP-digoxigenin nick end labeling
  • FIG. 89 depicts the design of siRNAs against apoptosis-specific eIF-5A.
  • the siRNAs have the SEQ ID NO: 45, 48, 51, 54 and 56.
  • the full-length nucleotide sequence is shown in SEQ ID NO: 29.
  • FIG. 90 shows the results of an experiment where siRNAs directed against apoptosis-specific eIF-5A provided for a reduction in NKkB activation in the presence of interferon gamma and LPS.
  • FIG. 91 shows the time course for PBMC experiments (see Example 18).
  • FIG. 92 shows a Western blot of a cell lysate from PBMCs collected from two donors over a time course.
  • the PBMCs were treated with PMA and subsequently stimulated with LPS to have an increased apoptosis-specific eIF-5A expression.
  • FIG. 93 shows that PBMCs treated with PMA and subsequently stimulated with LPS have an increased apoptosis-specific eIF-5A expression, which coincides with increased TNF production.
  • FIG. 94 demonstrates that PBMCs respond to LPS without PMA differentiation.
  • FIG. 95 shows that PBMCs transfected with apoptosis-specific eIF-5A siRNAs demonstrate suppression of expression of apoptosis-specific eIF-5A.
  • FIG. 96 shows that PBMCs transfected with apoptosis-specific eIF-5A siRNAs and stimulated with LPS produce less TNF than PBMCs not transfected with apoptosis-specific eIF-5A siRNAs.
  • FIG. 97 shows a western blot of cell lysate from HT-29 cells treated with or without gamma interferon.
  • FIG. 98 shows a western blot of cell lysate from HT-29 cells that were transfected with either control siRNA or apoptosis-specific eIF-5A siRNAs. This figure shows that siRNAs of apoptosis-specific eIF-5A inhibit expression of apoptosis-specific eIF-5A.
  • FIG. 99 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs have a reduced level of TNF production.
  • FIG. 100 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs exhibit a decreased in apoptosis as compared to control cells. Both control and siRNA-transfected cells were primed with interferon gamma and also treated with TNF- ⁇ .
  • FIG. 101 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs express less TLR4 protein than control cells.
  • FIG. 102 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs express less TNFR1 protein than control cells.
  • FIG. 103 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs express less iNOS protein than control cells.
  • FIG. 104 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs express less TLR4 mRNA than control cells.
  • FIG. 105 shows the time course for U937 treatments.
  • FIG. 106 shows that apoptosis-specific eIF-5A is upregulated with PMA in U937 cells.
  • FIG. 107 shows that apoptosis-specific eIF-5A is upregulated with LPS in U937 cells.
  • FIG. 108 shows that apoptosis-specific eIF-5A protein expression is still reduced after numerous hours following siRNA treatment.
  • FIG. 109 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction of TLR4.
  • FIG. 110 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with fewer glycosylated forms of the interferon gamma receptor in U937 cells.
  • FIG. 111 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction in TNFR1 in U937 cells.
  • FIG. 112 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction in LPS-induced TNF- ⁇ production in U937 cells.
  • FIG. 113 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction in LPS-induced IL-1 ⁇ production in U937 cells.
  • FIG. 114 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction in LPS-induced IL-8 production in U937 cells.
  • FIG. 115 shows that IL-6 production is independent of siRNA mediated down-regulation of apoptosis-specific eIF-5A in U937 cells.
  • eukaryotic initiation factor 5A (“eIF-5A”)
  • eIF-5A eukaryotic initiation factor 5A
  • DHS DHS
  • Apoptosis-specific eIF-5A is likely to be a suitable target for intervention in apoptosis-causing disease states since it appears to act at the level of post-transcriptional regulation of downstream effectors and transcription factors involved in the apoptotic pathway. Specifically, apoptosis-specific eIF-5A appears to selectively facilitate the translocation of mRNAs encoding downstream effectors and transcription factors of apoptosis from the nucleus to the cytoplasm, where they are subsequently translated. The ultimate decision to initiate apoptosis appears to stem from a complex interaction between internal and external pro- and anti-apoptotic signals. Lowe & Lin (2000) Carcinogenesis, 21, 485-495. Through its ability to facilitate the translation of downstream apoptosis effectors and transcription factors, apoptosis-specific eIF-5A appears to tip the balance between these signals in favor of apoptosis.
  • the present invention provides a method of suppressing or reducing apoptosis in a cell by administering an agent that inhibits or reduces expression of either apoptosis-specific eIF-5A or DHS.
  • an agent that inhibits or reduces expression of either apoptosis-specific eIF-5A or DHS By reducing expression of DHS, there is less DHS protein to be available to activate apoptosis-specific eIF-5A.
  • One agent that can inhibit or reduce expression of apoptosis-specific eIF-5A or DHS are antisense oligonucleotides of apoptosis-specific eIF-5A or DHS.
  • Antisense oligonucleotides have been successfully used to accomplish both in vitro as well as in vivo gene-specific suppression.
  • Antisense oligonucleotides are short, synthetic strands of DNA (or DNA analogs), RNA (or RNA analogs), or DNA/RNA hybrids that are antisense (or complimentary) to a specific DNA or RNA target.
  • Antisense oligonucleotides are designed to block expression of the protein encoded by the DNA or RNA target by binding to the target mRNA and halting expression at the level of transcription, translation, or splicing.
  • antisense oligonucleotides By using modified backbones that resist degradation (Blake et al., 1985), such as replacement of the phosphodiester bonds in the oligonucleotides with phosphorothioate linkages to retard nuclease degradation (Matzura and Eckstein, 1968), antisense oligonucleotides have been used successfully both in cell cultures and animal models of disease (Hogrefe, 1999). Other modifications to the antisense oligonucleotide to render the oligonucleotide more stable and resistant to degradation are known and understand by one skilled in the art.
  • Antisense oligonucleotide as used herein encompasses double stranded or single stranded DNA, double stranded or single stranded RNA, DNA/RNA hybrids, DNA and RNA analogs, and oligonucleotides having base, sugar, or backbone modifications.
  • the oligonucleotides may be modified by methods known in the art to increase stability, increase resistance to nuclease degradation or the like. These modifications are known in the art and include, but are not limited to modifying the backbone of the oligonucleotide, modifying the sugar moieties, or modifying the base.
  • the antisense oligonucleotides of the present invention have a nucleotide sequence encoding a portion or the entire coding sequence of an apoptosis-specific eIF-5A polypeptide or a DHS polypeptide.
  • the inventors have transfected various cell lines with antisense nucleotides encoding a portion of an apoptosis-specific eIF-5A polypeptide as described below and measured the number of cells undergoing apoptosis.
  • the cell populations that were transfected with the antisense oligonucleotides showed a decrease in the number of cells undergoing apoptosis as compared to like cell populations not having been transfected with the antisense oligos.
  • FIGS. 54-58 show a decrease in the percentage of cells undergoing apoptosis in the cells having being treated with antisense apoptosis-specific eIF-5A oligonucleotides as compared to cells not having been transfected with the antisense apoptosis-specific eIF-5A oligonucleotides.
  • the present invention contemplates the use of many suitable nucleic acid sequences encoding an apoptosis-specific eIF-5A polypeptide or DHS polypeptide.
  • the present invention provides antisense oligonucleotides of the following apoptosis-specific eIF-5A nucleic acid sequences (SEQ ID NOS: 1, 3, 4, 5, 11, 12, 15, 16, 19, 20, and 21) and DHS sequences (SEQ ID NOS:6, 7, 8).
  • Antisense oligonucleotides of the present invention need not be the entire length of the provided SEQ ID NOs. They need only be long enough to be able to bind to the mRNA and inhibit expression of such mRNA. “Inhibition or reduction of expression” or “suppression of expression” refers to the absence or detectable decrease in the level of protein and/or mRNA product from a target gene, such as apoptosis-specific eIF-5A.
  • antisense oligonucleotides of apoptosis-specific eIF-5A that do not comprise the entire coding sequence are antisense oligonucleotides of apoptosis-specific eIF-5A having the following SEQ ID NO: 35, 37, and 39.
  • Antisense oligonucleotide of apoptosis-specific eIF-5A includes oligonucleotides having substantial sequence identity or substantial homology to apoptosis-specific eIF-5A. Additional antisense oligonucleotides of apoptosis-specific eIF-5A of the present invention include those that have substantial sequence identity to those enumerated above (i.e. 90% homology) or those having sequences that hybridize under highly stringent conditions to the enumerated SEQ ID NOs. As used herein, the term “substantial sequence identity” or “substantial homology” is used to indicate that a sequence exhibits substantial structural or functional equivalence with another sequence.
  • any structural or functional differences between sequences having substantial sequence identity or substantial homology will be de minimus; that is, they will not affect the ability of the sequence to function as indicated in the desired application. Differences may be due to inherent variations in codon usage among different species, for example. Structural differences are considered de minimus if there is a significant amount of sequence overlap or similarity between two or more different sequences or if the different sequences exhibit similar physical characteristics even if the sequences differ in length or structure. Such characteristics include, for example, the ability to hybridize under defined conditions, or in the case of proteins, immunological crossreactivity, similar enzymatic activity, etc. The skilled practitioner can readily determine each of these characteristics by art known methods.
  • two nucleotide sequences are “substantially complementary” if the sequences have at least about 70 percent or greater, more preferably 80 percent or greater, even more preferably about 90 percent or greater, and most preferably about 95 percent or greater sequence similarity between them.
  • Two amino acid sequences are substantially homologous if they have at least 70%, more preferably at least 80%, even more preferably at least 90%, and most preferably at least 95% similarity between the active, or functionally relevant, portions of the polypeptides.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • at least 30%, 40%, 50%, 60%, 70%, 80%, or 90% or more of the length of a reference sequence is aligned for comparison purposes.
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • the nucleic acid and protein sequences of the present invention can further be used as a “query sequence” to perform a search against sequence databases to, for example, identify other family members or related sequences.
  • Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol Biol. 215:403-10.
  • BLAST nucleotide searches can be performed with the NBLAST program.
  • BLAST protein searches can be performed with the XBLAST program to obtain amino acid sequences homologous to the proteins of the invention.
  • Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25(17):3389-3402.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • apoptosis-specific eIF-5A includes functional derivatives thereof.
  • the term “functional derivative” of a nucleic acid is used herein to mean a homolog or analog of the gene or nucleotide sequence. A functional derivative may retain the function of the given gene, which permits its utility in accordance with the invention.
  • “Functional derivatives” of the apoptosis-specific eIF-5A polypeptide or functional derivatives of antisense oligonucleotides of apoptosis-specific eIF-5A as described herein are fragments, variants, analogs, or chemical derivatives of apoptosis-specific eIF-5A that retain Apoptosis-specific eIF-5A activity or immunological cross reactivity with an antibody specific for apoptosis-specific eIF-5A.
  • a fragment of the apoptosis-specific eIF-5A polypeptide refers to any subset of the molecule.
  • Functional variants can also contain substitutions of similar amino acids that result in no change or an insignificant change in function.
  • Amino acids that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham et al. (1989) Science 244:1081-1085). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity such as kinase activity or in assays such as an in vitro proliferative activity. Sites that are critical for binding partner/substrate binding can also be determined by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling (Smith et al. (1992) J. Mol. Biol. 224:899-904; de Vos et al. (1992) Science 255:306-312).
  • a “variant” refers to a molecule substantially similar to either the entire gene or a fragment thereof, such as a nucleotide substitution variant having one or more substituted nucleotides, but which maintains the ability to hybridize with the particular gene or to encode mRNA transcript which hybridizes with the native DNA.
  • a “homolog” refers to a fragment or variant sequence from a different animal genus or species.
  • An “analog” refers to a non-natural molecule substantially similar to or functioning in relation to the entire molecule, a variant or a fragment thereof.
  • Variant peptides include naturally occurring variants as well as those manufactured by methods well known in the art. Such variants can readily be identified/made using molecular techniques and the sequence information disclosed herein. Further, such variants can readily be distinguished from other proteins based on sequence and/or structural homology to the eIF-5A or DHS proteins of the present invention. The degree of homology/identity present will be based primarily on whether the protein is a functional variant or non-functional variant, the amount of divergence present in the paralog family and the evolutionary distance between the orthologs.
  • Non-naturally occurring variants of the eIF-5A or DHS polynucleotides, antisense oligonucleotides, or proteins of the present invention can readily be generated using recombinant techniques.
  • Such variants include, but are not limited to deletions, additions and substitutions in the nucleotide or amino acid sequence.
  • one class of substitutions are conserved amino acid substitutions.
  • Such substitutions are those that substitute a given amino acid in a protein by another amino acid of like characteristics.
  • conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu, and Ile; interchange of the hydroxyl residues Ser and Thr; exchange of the acidic residues Asp and Glu; substitution between the amide residues Asn and Gln; exchange of the basic residues Lys and Arg; and replacements among the aromatic residues Phe and Tyr.
  • Guidance concerning which amino acid changes are likely to be phenotypically silent are found in Bowie et al., Science 247:1306-1310 (1990).
  • hybridization is generally used to mean hybridization of nucleic acids at appropriate conditions of stringency as would be readily evident to those skilled in the art depending upon the nature of the probe sequence and target sequences. Conditions of hybridization and washing are well known in the art, and the adjustment of conditions depending upon the desired stringency by varying incubation time, temperature and/or ionic strength of the solution are readily accomplished. See, e.g. Sambrook, J. et al., Molecular Cloning: A Laboratory Manual, 2 nd edition, Cold Spring Harbour Press, Cold Spring Harbor, N.Y., 1989.
  • the choice of conditions is dictated by the length of the sequences being hybridized, in particular, the length of the probe sequence, the relative G-C content of the nucleic acids and the amount of mismatches to be permitted.
  • Low stringency conditions are preferred when partial hybridization between strands that have lesser degrees of complementarity is desired.
  • high stringency conditions are preferred.
  • High stringency conditions means that the hybridization solution contains 6 ⁇ S.S.C., 0.01 M EDTA, 1 ⁇ Denhardt's solution and 0.5% SDS.
  • Hybridization is carried out at about 68° C. for about 3 to 4 hours for fragments of cloned DNA and for about 12 to 16 hours for total eucaryotic DNA.
  • the temperature of hybridization is reduced to about 42° C. below the melting temperature (T m ) of the duplex.
  • T m melting temperature
  • hybridizes to a corresponding portion of a DNA or RNA molecule means that the molecule that hybridizes, e.g., oligonucleotide, polynucleotide, or any nucleotide sequence (in sense or antisense orientation) recognizes and hybridizes to a sequence in another nucleic acid molecule that is of approximately the same size and has enough sequence similarity thereto to effect hybridization under appropriate conditions. For example, a 100 nucleotide long sense molecule will recognize and hybridize to an approximately 100 nucleotide portion of a nucleotide sequence, so long as there is about 70% or more sequence similarity between the two sequences.
  • the size of the “corresponding portion” will allow for some mismatches in hybridization such that the “corresponding portion” may be smaller or larger than the molecule which hybridizes to it, for example 20-30% larger or smaller, preferably no more than about 12-15% larger or smaller.
  • amino acids that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham et al., Science 244:1081-1085 (1989)). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity or in assays.
  • the present invention also provides other agents that can inhibit or reduce expression of apoptosis-specific eIF-5A or DHS.
  • One such agent includes small inhibitory RNAs (“siRNA”).
  • siRNA technology has been emerging as a viable alternative to antisense oligonucleotides since lower concentrations are required to achieve levels of suppression that are equivalent or superior to those achieved with antisense oligonucleotides (Thompson, 2002).
  • Long double-stranded RNAs have been used to silence the expression of specific genes in a variety of organisms such as plants, nematodes, and fruit flies.
  • RNA-induced silencing complex An RNase-III family enzyme called Dicer processes these long double stranded RNAs into 21-23 nucleotide small interfering RNAs which are then incorporated into an RNA-induced silencing complex (RISC). Unwinding of the siRNA activates RISC and allows the single-stranded siRNA to guide the complex to the endogenous mRNA by base pairing. Recognition of the endogenous mRNA by RISC results in its cleavage and consequently makes it unavailable for translation. Introduction of long double stranded RNA into mammalian cells results in a potent antiviral response, which can be bypassed by use of siRNAs. (Elbashir et al., 2001). siRNA has been widely used in cell cultures and routinely achieves a reduction in specific gene expression of 90% or more.
  • siRNAs have also been gaining popularity in inhibiting gene expression in animal models of disease.
  • An siRNA against Fas, a receptor in the TNF family, injected hydrodynamically into the tail vein of mice was able to transfect greater than 80% of hepatocytes and decrease Fas expression in the liver by 90% for up to 10 days after the last injection (Song et al., 2003).
  • the Fas siRNA was also able to protect mice from liver fibrosis and fulminant hepatitis.
  • siRNA has the potential to be a very potent drug for the inhibition of specific gene expression in vitro in light of their long-lasting effectiveness in cell cultures and their ability to transfect cells in vivo and their resistance to degradation in serum in vivo (Bertrand et al., 2002) in vivo.
  • the present inventors have transfected cells with siRNAs of apoptosis-specific eIF-5A and studied the effects on expression of apoptosis-specific eIF-5A.
  • FIG. 64 shows that cells transfected with apoptosis-specific eIF-5A siRNA produced less apoptosis-specific eIF-5A protein.
  • FIGS. 65-67 show that cell populations transfected with apoptosis-specific eIF-5A siRNAs have a lower percentage of cells undergoing apoptosis after exposure to amptothecin and TNF- ⁇ as compared to cells not having been transfected with apoptosis-specific eIF-5A siRNAs.
  • one embodiment of the present invention provides for inhibiting expression of apoptosis-specific eIF-5A in cells by transfecting the cells with a vector comprising a siRNAs of apoptosis-specific eIF-5A.
  • siRNAs of apoptosis-specific eIF-5A include those that have SEQ ID NO: 31, 31, 32, and 33. Additional siRNAs include those that have substantial sequence identity to those enumerated (i.e. 90% homology) or those having sequences that hybridize under highly stringent conditions to the enumerated SEQ ID NOs. What is meant by substantial sequence identity and homology is described above with respect to antisense oligonucleotides of the present invention.
  • the term “siRNAs of apoptosis-specific eIF-5A” include functional variants or derivatives as described above with respect to antisense oligonucleotides of the present invention.
  • siRNA and expression constructs/vectors comprising siRNA are known by those skilled in the art.
  • U.S. applications 2004/106567 and 2004/0086884, which are herein incorporated by reference in their entirety, provide numerous expression constructs/vectors as well as delivery mechanism including viral vectors, non viral vectors, liposomal delivery vehicles, plasmid injection systems, artificial viral envelopes and poly-lysine conjugates to name a few.
  • regulatory sequences useful in expression constructs/vectors with antisense oligonucleotides or siRNA.
  • regulatory sequences may be a constitutive promoter, an inducible promoter, a tissue-specific promoter, or a combination thereof.
  • the methods and compositions of the present invention can be used to prevent or treat the following apoptosis-associated diseases and disorders: neurological/ neurodegenerative disorders (e.g., Alzheimer's, Parkinson's, Huntington's, Amyotrophic Lateral Sclerosis (Lou Gehrig's Disease), autoimmune disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus (SLE), multiple sclerosis), Duchenne Muscular Dystrophy (DMD), motor neuron disorders, ischemia, heart ischemia, chronic heart failure, stroke, infantile spinal muscular atrophy, cardiac arrest, renal failure, atopic dermatitis, sepsis and septic shock, AIDS, hepatitis, glaucoma
  • Glaucoma is a group of eye conditions arising from damage to the optic nerve that results in progressive blindness. Apoptosis has been shown to be a direct cause of this optic nerve damage.
  • IOP intra-ocular pressure
  • TNF- ⁇ tumor necrosis factor- ⁇
  • HVJ inactivated hemagglutinating virus of Japan
  • HVJ liposomes Intravitreal injection into mice of FITC-labeled antisense oligonucleotides encapsulated in HVJ liposomes resulted in high fluorescence within 44% of the cells in the ganglion layer which lasted three days while fluorescence with naked FITC-labeled antisense oligonucleotide disappeared after one day.
  • HVJ liposomes Intravitreal injection into mice of FITC-labeled antisense oligonucleotides encapsulated in HVJ liposomes
  • One method of preventing or reducing apoptosis of the present invention is directed to preventing or reducing apoptosis in cells and tissues of the eye, such as but not limited to, astrocytes, retinal ganglion, retinal glial cells and lamina cribosa. Death of retinal ganglion cells in glaucoma occurs by apoptosis and which leads to blindness.
  • providing a method of inhibiting or reducing apoptosis in retinal ganglion cells or by protecting retinal ganglion cells from degeneration by apoptosis provides a novel treatment for prevention of blindness due to glaucoma.
  • the present invention provides a method for preventing or inhibiting retinal ganglion cell death in a glaucomatous eye, by suppressing expression of apoptosis-specific eIF-5A. Inhibiting the expression of apoptosis-specific eIF-5A reduces apoptosis.
  • Apoptosis-specific eIF-5A is a powerful gene that appears to regulate the entire apoptotic process. Thus, controlling apoptosis in the optic nerve head indicates that blocking expression of apoptosis-specific eIF-5A provides a treatment for glaucoma.
  • Suppression of expression of apoptosis-specific eIF-5A is accomplished by administering an antisense oligonucleotides or a siRNA of human apoptosis-specific eIF-5A to cells of the eye such as, but not limited to lamina cribrosa, astrocytes, retinal ganglion, or retinal glial cells.
  • Antisense oligonucleotides and siRNAs are as defined above, i.e. have a nucleotide sequence encoding at least a portion of an apoptosis-specific eIF-5A polypeptide.
  • Exemplary antisense oligonucleotides useful in this aspect of the invention comprise SEQ ID NO:26 or 27 or oligonucleotides that bind to a sequence complementary to SEQ ID NO:26 or 27 under high stringency conditions and which inhibit expression of apoptosis-specific eIF-5A.
  • Another embodiment of the invention provides a method of suppressing expression of apoptosis-specific eIF-5A in lamina cribosa cells, astrocyte cells, retinal ganglion cells or retinal glial cells.
  • Antisense oligonucleotides or siRNAs such as but not limited to, SEQ ID NO:26 and 27, targeted against human apoptosis-specific eIF-5A are administered to lamina cribosa cells, astrocyte cells, retinal ganglion cells or retinal glial cells.
  • the cells may be of human origin.
  • eIF5A may also play a role in the immune response.
  • the present inventors have discovered that apoptosis-specific eIF-5A levels correlate with elevated levels of two cytokines (Interleukin 1-beta “IL-1 ⁇ ” and interleukin 18 “IL-18”) in ischemic heart tissue, thus further proving that apoptosis-specific eIF-5A is involved in cell death as it is present in ischemic heart tissue. This apoptosis-specific eIF-5A/interleukin correlation is not seen in non-ischemic heart tissue. See FIGS. 50 A-F and 51 .
  • eIF-5A2 proliferating eIF-5A
  • IL-1 ⁇ proliferating eIF-5A
  • apoptosis-specific eIF-5A The correlation of apoptosis-specific eIF-5A to these potent interleukins further suggests that the inflammation and apoptosis pathways in ischemia may be controlled via controlling levels of apoptosis-specific eIF-5A. Further evidence that apoptosis-specific eIF-5A is involved in the immune response is suggested by the fact that human peripheral blood mononuclear cells (PBMCs) normally express very low levels of eIF-5A, but upon stimulation with T-lymphocyte-specific stimuli expression of apoptosis-specific eIF-5A increases dramatically (Bevec et al., 1994). This suggests a role for apoptosis-specific eIF-5A in T-cell proliferation and/or activation.
  • PBMCs peripheral blood mononuclear cells
  • eIF-5A may be required as a nucleocytoplasmic shuttle for cytokine mRNAs.
  • the authors of the above referenced article also found elevated levels of eIF5A in the PBMCs of HIV-1 patients which may contribute to efficient HIV replication in these cells as eIF5A has been demonstrated to be a cellular binding factor for the HIV Rev protein and required for HIV replication (Ruhl et al., 1993).
  • Dendritic cells are antigen-presenting cells that sensitize helper and killer T cells to induce T cell-mediated immunity (Steinman, 1991). Immature dendritic cells lack the ability to stimulate T cells and require appropriate stimuli (i.e. inflammatory cytokines and/or microbial products) to mature into cells capable of activating T cells.
  • An inhibitor of deoxyhypusine synthase, the enzyme required to activate eIF5A was found to inhibit T lymphocyte activation by dendritic cells by preventing CD83 surface expression (Kruse et aL, 2000).
  • eIF5A may facilitate dendritic cell maturation by acting as a nucleocytoplasmic shuttle for CD83 mRNA.
  • the present invention relates to controlling the expression of apoptosis-specific eIF-5A to control the rate of dendritic cell maturation and PBMC activation, which in turn may control the rate of T cell-mediated immunity.
  • the present inventors studied the role of apoptosis-specific eIF-5A in the differentiation of monocytes into adherent macrophages using the U-937 cell line, as U-937 is known to express eIF-5A mRNA (Bevec et al., 1994).
  • U-937 is a human monocyte cell line that grows in suspension and will become adherent and differentiate into macrophages upon stimulation with PMA.
  • one aspect of the invention provides for a method of inhibiting or delaying maturation of macrophages to inhibit or reduce the production of cytokines. This method involves providing an agent that is capable of reducing the expression of either DHS or apoptosis-specific eIF-5A. By reducing or eliminating expression of DHS, apoptosis-specific eIF-5A activation will be reduced or eliminated.
  • apoptosis-specific eIF-5A is upregulated during monocyte differentiation and TNF- ⁇ secretion, it is believed that it is necessary for these events to occur. Thus, by reducing or eliminating activation of apoptosis-specific eIF-5A or by directly reducing or eliminating apoptosis-specific eIF-5A expression, monocyte differentiation and TNF- ⁇ secretion can be reduced or eliminated.
  • Any agent capable of reducing the expression of DHS or apoptosis-specific eIF-5A may be used and includes, but is not limited to antisense oligonucleotides or siRNAs as described herein.
  • the present inventors have studied the ability of human apoptosis-specific eIF-5A to promote translation of cytokines by acting as a nucleocytoplasmic shuttle for cytokine mRNAs in vitro using a cell line known to predictably produce cytokine(s) in response to a specific stimulus. Some recent studies have found that human liver cell lines can respond to cytokine stimulation by inducing production of other cytokines. HepG2 is a well characterized human hepatocellular carcinoma cell line found to be sensitive to cytokines.
  • HepG2 cells In response to IL-1 ⁇ , HepG2 cells rapidly produce TNF- ⁇ mRNA and protein in a dose-dependent manner (Frede et al., 1996; Rowell et al., 1997; Wordemann et al., 1998). Thus, HepG2 cells were used as a model system to study the regulation of TNF- ⁇ production. The present inventors have shown that inhibition of human apoptosis-specific eIF-5A expression in HepG2 cells caused the cells to produce less TNF- ⁇ after having been transfected with antisense oligonucleotide of directed toward apoptosis factor 5A.
  • one embodiment of the present invention provides a method for reducing levels of a cytokine.
  • the method involves administering an agent capable of reducing expression of apoptosis factor 5A1. Reducing expression of apoptosis factor 5A1 also reduces expression of the cytokine and thus leads to a decreased amount of the cytokine produced by cell.
  • the cytokine is a preferably a pro-inflammatory cytokine, including, but not limited to IL-1, IL-18, IL-6 and TNF- ⁇ .
  • Antisense oligonucleotides are as discussed above.
  • Exemplary antisense oligonucleotides of human apoptosis-specific eIF-5A are selected from the group consisting of SEQ ID NO: 35, 37, and 39 or is an antisense nucleotide that hybridizes under highly stringent conditions to a sequence selected from the group consisting of SEQ ID NO: 35, 37, and 39.
  • An agent may also comprise a siRNA of human apoptosis-specific eIF-5A and are as discussed above.
  • exemplary siRNAs have a sequence selected from the group consisting of SEQ ID NO: 30, 31, 32 and 33 or is a siRNA that hybridizes under highly stringent conditions to a sequence selected from the group consisting of SEQ ID NO: 30, 31, 32 and 33.
  • FIGS. 65-67 show that cells transfected with human apoptosis-specific eIF-5A siRNAs have a lower percentage of cells undergoing apoptosis after exposure to amptothecin and TNF- ⁇ .
  • the present invention is also directed to a method for reducing the expression of p53.
  • This method involves administering an agent capable of reducing expression of apoptosis factor 5A, such as the antisense oligonucleotides or the siRNAs described above. Reducing expression of apoptosis-specific eIF-5A reduces expression of p53 as shown in FIG. 52 and example 10.
  • the present invention is also directed to a method for increasing the expression of Bcl-2.
  • This method entails administering an agent capable of reducing expression of human apoptosis-specific eIF-5A.
  • Preferred agents include antisense oligonucleotides and siRNAs described above. Reducing expression of apoptosis-specific eIF-5A increases expression of Bcl-2 as shown in FIG. 63 and example 13.
  • FIG. 63 shows that cells transfected with apoptosis-specific eIF-5A siRNA produced less apoptosis-specific eIF-5A protein and in addition, produced more Bcl-2 protein. A decrease in apoptosis-specific eIF-5A expression correlates with an increase in BCL-2 expression.
  • the present invention also provides a method for reducing levels of TNF-alpha in a patient in need thereof comprising administering to said patient either antisense oligonucleotide or siRNAs of apoptosis-specific eIF-5A as described above.
  • administering to said patient either antisense oligonucleotide or siRNAs of apoptosis-specific eIF-5A as described above.
  • FIG. 69 and example 14 cells transfected with antisense apoptosis-specific eIF-5A oligonucleotides of the present invention produced less TNF- ⁇ after induction with IL-1than cells not transfected with such antisense oligonucleotides.
  • the present invention provides a method of treating pathological conditions characterized by an increased IL-1, TNF-alpha, IL-61 or IL-18 level comprising administering to a mammal having said pathological condition, agents to reduce expression of apoptosis-specific eIF-5A as described above (antisense oligonucleotides and siRNA).
  • IL-1, TNF-alpha, or Il-6 levels include, but are not limited to arthritis-rheumatoid and osteo arthritis, asthma, allergies, arterial inflammation, crohn's disease, inflammatory bowel disease, (ibd), ulcerative colitis, coronary heart disease, cystic fibrosis, diabetes, lupus, multiple sclerosis, graves disease, periodontitis, glaucoma & macular degeneration, ocular surface diseases including keratoconus, organ ischemia-heart, kidney, repurfusion injury, sepsis, multiple myeloma, organ transplant rejection, psoriasis and eczema.
  • reducing expression of apoptosis-specific eIF-5A with the antisense oligonucleotids, siRNAs and methods of the present invention may provide relief from these pathological conditions.
  • the present invention also provides a method of delivering siRNA to mammalian lung cells in vivo.
  • siRNAs directed against apoptosis-specific eIF-5A were administered intranasally (mixed with water) to mice.
  • lipopolysaccharide (LPS) was administered intranasally to the mice.
  • the right lung was removed and myeloperoxidase was measured.
  • the mouse apoptosis-specific eIF-5A siRNA suppressed myeloperoxidase by nearly 90% as compared to the control siRNA. In the study, there were 5 mice in each group.
  • MPO Myeloperoxidase
  • the myeloperoxidase is an enzyme that uses hydrogen peroxidase to convert chloride to hypochlorous acid. The hypochlorous acid reacts with and destroys bacteria. Myeloperoxidase is also produced when arteries are inflamed.
  • myeloperoxidase is associated with neutrophils and the inflammation response.
  • the present inventors have shown that by down regulating apoptosis-specific eIF-5A with siRNAs shows a marked decrease in the myeloperoxidase in lung tissue after exposed to LPS (which normally produces an inflarnmatory response involving the production of myeloperoxidase).
  • LPS which normally produces an inflarnmatory response involving the production of myeloperoxidase.
  • siRNAs against apoptosis-specific eIF-5A can decrease or suppress the amount of myeloperoxidase in lung tissue and thus decrease or suppress the inflammation response.
  • LPS is a macromolecular cell surface antigen of bacteria that when applied in vivo triggers a network of inflammatory responses. Intranasally delivering LPS causes an increase in the number of neutrophils in the lungs. One of the primary events is the activation of mononuclear phagocytes through a receptor-mediated process, leading to the release of a number of cytokines, including TNF- ⁇ . In turn, the increased adherence of neutrophils to endothelial cells induced by TNF- ⁇ leads to massive infiltration in the pulmonary space.
  • siRNAs against apoptosis-specific eIF-5A an suppress the production of myeloperoxidase (i.e. part of the inflammation response).
  • the inventors have also shown that it is possible to deliver siRNAs in vivo to lung tissue by simple intranasal delivery. The siRNAs were mixed only in water. This presents a major breakthrough and discovery as others skilled in the art have attempted to design acceptable delivery methods for siRNA.
  • MPO levels are a critical predictor of heart attacks and cytokine-induced inflammation caused by autoimmune disorders. This ability to decrease or suppress the inflammation response may serve useful in treating inflammation related disorders such as auto immune disorders. In addition, the ability to lower MPO could be a means of protecting patients from ischemic events and heart attacks.
  • mice were similarly treated with siRNAs directed against apoptosis-specific eIF-5A.
  • Lipopolysaccharide (LPS) was administered to the mice to induce inflammation and an immune system response.
  • LPS kills thymocytes, which are important immune system precursor cells created in the thymus to fend off infection.
  • using the siRNAs directed against apoptosis-specific eIF-5A allowed approximately 90% survivability of the thymocytes in the presence of LPS.
  • thymocytes are destroyed, since they are precursors to T cells, the body's natural immunity is compromised by not being able to produce T cells and thus can't ward off bacterial infections and such.
  • using the siRNAs against apoptosis-specific eIF-5A can be used to reduce inflammation (as shown by a lower level of MPO in the first example) without destroying the body's natural immune defense system.
  • NFk beta NFk beta
  • NFkB NFk beta
  • COX-2 NFk beta
  • NFkB NFk beta resides in the cytoplasm of the cell and is bound to its inhibitor. Injurious and inflammatory stimuli release NFkB from the inhibitor. NFkB moves into the nucleus and activates the genes responsible for expressing COX-2.
  • inflammation can be reduced.
  • HT-29 cells human epithelial cells
  • siRNA targeted at apoptosis-specific eIF-5A Inflammation was then induced by NFkB by addition of TNF or interferon gamma and LPS for one hour.
  • the results of this experiment show that inhibiting the expression of apoptosis-specific eIF-5A with siRNAs provided for a reduction in the levels of NFKB that were activated by the gamma interferon and LPS. See FIG. 74 .
  • One embodiment of the present invention provides methods of inhibition expression of endogenous apoptosis-specific eIF-5A in a cell. Inhibiting expression is preferably carried out by the use of antisense polynucleotides or siRNAs of apoptosis-specific eIF-5A of the present invention described previously.
  • expression of endogenous apoptosis-specific eIF-5A occurs various effects on the cell result. For example, a reduction in expression of the various proteins, factors, receptors, cytokines are seen: p53; pro-inflammatory cytokines (See FIG. 112, 113 , 114 ); myeloperoxidase; active NFk beta; TLR4 (See FIG. 109 ); TNFR-1 (See FIG. 111 ) and iNOS.
  • a reduction in expression of endogenous apoptosis-specific eIF-5A increases levels expression of Bcl-2 in the cell.
  • the antisense nucleic acids siRNAs of the present invention where used in an animal for the purpose of prophylaxis or treatment, will be administered in the form of a composition additionally comprising a pharmaceutically acceptable carrier.
  • suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • Pharmaceutically acceptable carriers can further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the binding proteins.
  • the compositions of the injection can, as is well known in the art, be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the mammal.
  • compositions of this invention can be in a variety of forms. These include, for example, solid, semi-solid and liquid dosage forms, such as tablets, pills, powders, liquid solutions, dispersions or suspensions, liposomes, suppositories, injectable and infusible solutions.
  • solid, semi-solid and liquid dosage forms such as tablets, pills, powders, liquid solutions, dispersions or suspensions, liposomes, suppositories, injectable and infusible solutions.
  • the preferred form depends on the intended mode of administration and therapeutic application.
  • compositions can be prepared in a manner well known in the pharmaceutical art.
  • the active ingredient will usually be mixed with a carrier, or diluted by a carrier, and/or enclosed within a carrier which can, for example, be in the form of a capsule, sachet, paper or other container.
  • a carrier which can, for example, be in the form of a capsule, sachet, paper or other container.
  • the carrier serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, excipient or medium for the active ingredient.
  • the composition can be in the form of tablets, lozenges, sachets, cachets, elixirs, suspensions, aerosols (as a solid or in a liquid medium), ointments containing for example up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, injection solutions, suspensions, sterile packaged powders and as a topical patch.
  • the degree of apoptosis was determined by DNA laddering. Genomic DNA was isolated from dispersed corpus luteal cells or from excised corpus luteum tissue using the QIAamp DNA Blood Kit (Qiagen) according to the manufacturer's instructions. Corpus luteum tissue was excised before the induction of apoptosis by treatment with PGF-2 ⁇ , 1 hour and 24 hours after induction of apoptosis.
  • the isolated DNA was end-labeled by incubating 500 ng of DNA with 0.2 ⁇ Ci[ ⁇ - 32 P]dCTP, 1 mM Tris, 0.5 mM EDTA, 3 units of Klenow enzyme, and 0.2 pM each of dATP, dGTP, and dTTP at room temperature for 30 minutes. Unincorporated nucleotides were removed by passing the sample through a 1 ml Sepadex G-50 column according to Sambrook et al. The samples were then resolved by Tris-acetate-EDTA (1.8%) gel electrophoresis. The gel was dried for 30 minutes at room temperature under vacuum and exposed to x-ray film at ⁇ 80° C. for 24 hours.
  • the degree of apoptosis in superovulated rat corpus lutea was examined either 0, 1, or 24 hours after injection with PGF-2 ⁇ .
  • the ovaries were removed without PGF-2 ⁇ injection.
  • Laddering of low molecular weight DNA fragments reflecting nuclease activity associated with apoptosis is not evident in control corpus luteum tissue excised before treatment with PGF-2 ⁇ , but is discernible within 1 hour after induction of apoptosis and is pronounced by 24 hours after induction of apoptosis, which is shown in FIG. 16 .
  • the top panel is an autoradiograph of the Northern blot probed with the 32 P-dCTP-labeled 3′-untranslated region of rat corpus luteum apoptosis-specific DHS cDNA.
  • the lower panel is the ethidium bromide stained gel of total RNA. Each lane contains 10 ⁇ g RNA. The data indicate that there is down-regulation of apoptosis-specific eIF-5A transcript following serum withdrawal.
  • Control cells from rats treated with saline were dispersed with collagenase and incubated for 1 hour and a further 5 hours in glutamine only (lane 1).
  • Five hundred nanograms of DNA from each sample was labeled with [ ⁇ - 32 P]-dCTP using klenow enzyme, separated on a 1.8% agarose gel, and exposed to film for 24 hours. Results are shown in FIG. 18 .
  • superovulated rats were injected subcutaneously with 1 mg/100 g body weight of spermidine, delivered in three equal doses of 0.333 mg/100 g body weight, 24, 12, and 2 hours prior to a subcutaneous injection with 500 ⁇ g PGF-2 ⁇ .
  • Control rats were divided into three sets: no injections, three injections of spermidine but no PGF-2 ⁇ ; and three injections with an equivalent volume of saline prior to PGF-2 ⁇ treatment.
  • Ovaries were removed front the rats either 1 hour and 35 minutes or 3 hours and 45 minutes after prostaglandin treatment and used for the isolation of DNA.
  • guanidinium buffer 4 M guanidinium isothiocyanate, 2.5 mM NaOAc pH 8.5, 0.8% ⁇ -mercaptoethanol
  • the pelleted RNA was rinsed with 75% ethanol, resuspended in 600 ml DEPC-treated water and the RNA precipitated at ⁇ 70° C. with 1.5 ml 95% ethanol and 60 ml of 3M NaOAc.
  • Genomic DNA was isolated from extracted corpus luteum tissue or dispersed corpus luteal cells using the QIAamp DNA Blood Kit (Qiagen) according to the manufacturer's instructions.
  • the DNA was end-labeled by incubating 500 ng of DNA with 0.2 ⁇ Ci[ ⁇ - 32 P]dCTP, 1 mM Tris, 0.5 mM EDTA, 3 units of Klenow enzyme, and 0.2 pM each of dATP, dGTP, and dTTP, at room temperature for 30 minutes. Unincorporated nucleotides were removed by passing the sample through a 1-ml Sephadex G-50 column according to the method described by Maniatis et al. The samples were then resolved by Tris-acetate-EDTA (2%) gel electrophoresis. The gel was dried for 30 minutes at room temperature under vacuum and exposed to x-ray film at ⁇ 80° C. for 24 hours.
  • RNA isolated from rat corpus luteum at various stages of apoptosis were separated on 1% denatured formaldehyde agarose gels and immobilized on nylon membranes.
  • the full-length rat apoptosis-specific eIF-5A cDNA (SEQ ID NO:1) labeled with 32 P-dCTP using a random primer kit (Boehringer) was used to probe the membranes 7 ⁇ 10 7 .
  • full length rat DHS cDNA (SEQ ID NO:6) labeled with 32 P-dCTP using a random primer kit (Boehringer) was used to probe the membranes (7 ⁇ 10 7 cpm).
  • the membranes were washed once with 1 ⁇ SSC, 0.1% SDS at room temperature and three times with 0.2 ⁇ SSC, 0.1% SDS at 65° C.
  • the membranes were dried and exposed to X-ray film overnight at ⁇ 70° C.
  • apoptosis-specific eIF-5A and DHS are both upregulated in apoptosing corpus luteum tissue.
  • Expression of apoptosis-specific eIF-5A is significantly enhanced after induction of apoptosis by treatment with PGF-2 ⁇ —low at time zero, increased substantially within 1 hour of treatment, increased still more within 8 hours of treatment and increased slightly within 24 hours of treatment ( FIG. 14 ).
  • Expression of DHS was low at time zero, increased substantially within 1 hour of treatment, increased still more within 8 hours of treatment and increased again slightly within 24 hours of treatment ( FIG. 15 ).
  • a partial-length apoptosis-specific eIF-5A sequence (SEQ ID NO:11) corresponding to the 3′ end of the gene was generated from apoptosing rat corpus luteum RNA template by RT-PCR using a pair of oligonucleotide primers designed from yeast, fungal and human apoptosis-specific eIF-5A sequences.
  • the upstream primer used to isolate the 3′end of the rat apoptosis-specific eIF-5A gene is a 20 nucleotide degenerate primer: 5′ TCSAARACHGGNAAGCAYGG 3′ (SEQ ID NO:9), wherein S is selected from C and G; R is selected from A and G; H is selected from A, T, and C; Y is selected from C and T; and N is any nucleic acid.
  • the downstream primer used to isolate the 3′end of the rat eIF-5A gene contains 42 nucleotides: 5′ GCGAAGCTTCCATGG CTCGAGTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTT 3′ (SEQ ID NO: 10).
  • RT-PCR reverse transcriptase polymerase chain reaction
  • a partial-length apoptosis-specific eIF-5A sequence (SEQ ID NO:15) corresponding to the 5′ end of the gene and overlapping with the 3′ end was generated from apoptosing rat corpus luteum RNA template by RT-PCR.
  • the 5′ primer is a 24-mer having the sequence, 5′ CAGGTCTAGAGTTGGAATCGAAGC 3′ (SEQ ID NO:13), that was designed from human eIF-5A sequences.
  • the 3′ primer is a 30-mer having the sequence, 5′ ATATCTCGAGCCTT GATTGCAACAGCTGCC 3′ (SEQ ID NO:14) that was designed according to the 3′ end RT-PCR fragment.
  • RT-PCR reverse transcriptase-polymerase chain reaction
  • the sequences of the 3′ and 5′ ends of the rat apoptosis-specific eIF-5A overlapped and gave rise to the full-length cDNA sequence (SEQ ID NO:1). This full-length sequence was aligned and compared with sequences in the GeneBank data base. See FIGS. 1-2 .
  • the cDNA clone encodes a 154 amino acid polypeptide (SEQ ID NO:2) having a calculated molecular mass of 16.8 KDa.
  • the nucleotide sequence, SEQ ID NO:1, for the full length cDNA of the rat apoptosis-specific corpus luteum eIF-5A gene obtained by RT-PCR is depicted in FIG. 3 and the corresponding derived amino acid sequence is SEQ ID NO:9.
  • the derived full-length amino acid sequence of eIF-5A was aligned with human and mouse eIF-5a sequences. See FIG. 7-9 .
  • a partial-length DHS sequence (SEQ ID NO:6) corresponding to the 3′ end of the gene was generated from apoptosing rat corpus luteum RNA template by RT-PCR using a pair of oligonucleotide primers designed from a human DHS sequence.
  • the 5′ primer is a 20-mer having the sequence, 5′ GTCTGTGTATTATTGGGCCC 3′ (SEQ ID NO. 17); the 3′ primer is a 42-mer having the sequence, 5′ GCGAAGCTTCCATGGC TCGAGTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTTT 3′ (SEQ ID NO:18).
  • RT-PCR reverse transcriptase polymerase chain reaction was carried out. Briefly, using 5 mg of the downstream primer, a first strand of cDNA was synthesized. The first strand was then used as a template in a RT-PCR using both the upstream and downstream primers.
  • Genomic DNA for southern blotting was isolated from excised rat ovaries. Approximately 100 mg of ovary tissue was divided into small pieces and placed into a 15 ml tube. The tissue was washed twice with 1 ml of PBS by gently shaking the tissue suspension and then removing the PBS using a pipette. The tissue was resuspended in 2.06 ml of DNA-buffer (0.2 M Tris-HCl pH 8.0 and 0.1 mM EDTA) and 240 ⁇ l of 10% SDS and 100 ⁇ l of proteinase K (Boehringer Manheim; 10 mg/ml) was added. The tissue was placed in a shaking water bath at 45° C. overnight.
  • DNA-buffer 0.2 M Tris-HCl pH 8.0 and 0.1 mM EDTA
  • tissue suspension was incubated in a water-bath at 45° C. for an additional 4 hours. After the incubation the tissue suspension was extracted once with an equal volume of phenol:chloroform:iso-amyl alcohol (25:24:1) and once with an equal volume of chloroform:iso-amyl alcohol (24:1). Following the extractions 1/10th volume of 3M sodium acetate (pH 5.2) and 2 volumes of ethanol were added. A glass pipette sealed and formed into a hook using a Bunsen burner was used to pull the DNA threads out of solution and to transfer the DNA into a clean microcentrifuge tube.
  • the DNA was washed once in 70% ethanol and air-dried for 10 minutes.
  • the DNA pellet was dissolved in 500 ⁇ l of 10 mM Tris-HCl (pH 8.0), 10 ⁇ l of RNase A (10 mg/ml) was added, and the DNA was incubated for 1 hour at 37° C.
  • the DNA was extracted once with phenol:chloroforrn:iso-amyl alcohol (25:24:1) and the DNA was precipitated by adding 1/10th volume of 3 M sodium acetate (pH 5.2) and 2 volumes of ethanol.
  • the DNA was pelleted by centrifugation for 10 minutes at 13,000 ⁇ g at 4° C.
  • the DNA pellet was washed once in 70% ethanol and dissolved in 200 ⁇ l 10 mM Tris-HCl (pH 8.0) by rotating the DNA at 4° C. overnight.
  • genomic DNA isolated from rat ovaries was digested with various restriction enzymes that either do not cut in the endogenous gene or cut only once. To achieve this, 10 ⁇ g genomic DNA, 20 ⁇ l 10 ⁇ reaction buffer and 100 U restriction enzyme were reacted for five to six hours in a total reaction volume of 200 ⁇ l. Digested DNA was loaded onto a 0.7% agarose gel and subjected to electrophoresis for 6 hours at 40 volts or overnight at 15 volts.
  • the gel was depurinated for 10 minutes in 0.2 N HCl followed by two 15-minute washes in denaturing solution (0.5 M NaOH, 1.5 M NaCl) and two 15 minute washes in neutralizing buffer (1.5 M NaCl, 0.5 M Tris-HCl pH 7.4).
  • the DNA was transferred to a nylon membrane, and the membrane was prehybridized in hybridization solution (40% formamide, 6 ⁇ SSC, 5 ⁇ Denhart's, solution (1 ⁇ Denhart's solution is 0.02% Ficoll, 0.02% PVP, and 0.02% BSA), 0.5% SDS, and 1.5 mg of denatured salmon sperm DNA).
  • a 700 bp PCR fragment of the 3′ UTR of rat eIF-5A cDNA (650 bp of 3′ UTR and 50 bp of coding) was labeled with [a-32P]-dCTP by random priming and added to the membrane at 1 ⁇ 106 cpm/ml.
  • a 606 bp PCR fragment of the rat DHS cDNA (450 bp coding and 156 bp 3′ UTR) was random prime labeled with [ ⁇ - 32 P]-dCTP and added at 1 ⁇ 10 6 cpm/ml to a second identical membrane.
  • the blots were hybridized overnight at 42° C. and then washed twice with 2 ⁇ SSC and 0.1% SDS at 42° C. and twice with 1 ⁇ SSC and 0.1% SDS at 42° C. The blots were then exposed to film for 3-10 days.
  • Rat corpus genomic DNA was cut with restriction enzymes as indicated on FIG. 20 and probed with 32 P-dCTP-labeled full-length eIF-5A cDNA. Hybridization under high stringency conditions revealed hybridization of the full-length cDNA probe to several restriction fragments for each restriction enzyme digested DNA sample, indicating the presence of several isoforms of eIF-5A.
  • EcoRV EcoRV
  • two restriction fragments of the apoptosis-specific isoform of eIF-5A were detectable in the Southern blot. The two fragments are indicated with double arrows in FIG. 20 .
  • the restriction fragment corresponding to the apoptosis-specific isoform of eIF-5A is indicated by a single arrow in the lanes labeled EcoRI and BamH1, restriction enzymes for which there are no cut sites within the open reading frame.
  • FIG. 21 shows a Southern blot of rat genomic DNA probed with 32 P-dCTP-labeled partial-length rat corpus luteum DHS cDNA.
  • the genomic DNA was cut with EcoRV, a restriction enzyme that does not cut the partial-length cDNA used as a probe. Two restriction fragments are evident indicating that there are two copies of the gene or that the gene contains an intron with an EcoRV site.
  • the present example demonstrates modulation of apoptosis apoptosis-specific eIF-5A (increasing apoptosis with apoptosis-specific eIF-5A in sense orientation)
  • COS-7 an African green monkey kidney fibroblast-like cell line transformed with a mutant of SV40 that codes for wild-type T antigen, was used for all transfection-based experiments.
  • COS-7 cells were cultured in Dulbecco's Modified Eagle's medium (DMEM) with 0.584 grams per liter of L-glutamine, 4.5 g of glucose per liter, and 0.37% sodium bicarbonate. The culture media was supplemented with 10% fetal bovine serum (FBS) and 100 units of penicillin/streptomycin. The cells were grown at 37° C. in a humidified environment of 5% CO 2 and 95% air.
  • DMEM Dulbecco's Modified Eagle's medium
  • FBS fetal bovine serum
  • the cells were subcultured every 3 to 4 days by detaching the adherent cells with a solution of 0.25% trypsin and 1 mM EDTA.
  • the detached cells were dispensed at a split ratio of 1:10 in a new culture dish with fresh media.
  • COS-7 cells to be used for isolation of RNA were grown in 150-mm tissue culture treated dishes (Coming). The cells were harvested by detaching them with a solution of trypsin-EDTA. The detached cells were collected in a centrifuge tube, and the cells were pelleted by centrifugation at 3000 rpm for 5 minutes. The supernatant was removed, and the cell pellet was flash-frozen in liquid nitrogen. RNA was isolated from the frozen cells using the GenElute Mammalian Total RNA Miniprep kit (Sigma) according to the manufacturer's instructions.
  • Recombinant plasmids carrying the full-length coding sequence of rat apoptosis-specific eIF-5A in the sense orientation and the 3′ untranslated region (UTR) of rat apoptosis-specific eIF-5A in the antisense orientation were constructed using the mammalian epitope tag expression vector, pHM6 (Roche Molecular Biochemicals), which is illustrated in FIG. 21 .
  • the vector contains the following: CMV promoter—human cytomegalovirus immediate-early promoter/enhancer; HA—nonapeptide epitope tag from influenza hemagglutinin; BGH pA—Bovine growth hormone polyadenylation signal; f1 ori—f1 origin; SV40 ori—SV40 early promoter and origin; Neomycin—Neomycin resistance (G418) gene; SV40 pA—SV40 polyadenylation signal; Col E1—ColE1 origin; Ampicillin—Ampicillin resistance gene.
  • the full-length coding sequence of rat apoptosis-specific eIF-5A and the 3′ UTR of rat apoptosis-specific eIF-5A were amplified by PCR from the original rat eIF-5A RT-PCR fragment in pBluescript (SEQ ID NO:1).
  • the primers used were as follows: Forward 5′ GCC AAGCTT AATGGCAGATGATTT GG 3′ (SEQ ID NO: 59) (Hind3) and Reverse 5′ CT GAATTC CAGT TATTTTGCCATGG 3′ (SEQ ID NO:60) (EcoR1).
  • the full-length rat apoptosis-specific eIF-5A PCR product isolated after agarose gel electrophoresis was 430 bp in length while the 3′ UTR rat apoptosis-specific eIF-5A PCR product was 697 bp in length. Both PCR products were subcloned into the Hind 3 and EcoR1 sites of pHM6 to create pHM6-full-length apoptosis-specific eIF-5A and pHM6-antisense 3′UTReIF-5A.
  • the full-length rat apoptosis-specific eIF-5A PCR product was subcloned in frame with the nonapeptide epitope tag from influenza hemagglutinin (HA) present upstream of the multiple cloning site to allow for detection of the recombinant protein using an anti-[HA]-peroxidase antibody.
  • HA hemagglutinin
  • Expression is driven by the human cytomegalovirus immediate-early promoter/enhancer to ensure high level expression in mammalian cell lines.
  • the plasmid also features a neomycin-resistance (G418) gene, which allows for selection of stable transfectants, and a SV40 early promoter and origin, which allows episomal replication in cells expressing SV40 large T antigen, such as COS-7.
  • G418 neomycin-resistance
  • SV40 early promoter and origin which allows episomal replication in cells expressing SV40 large T antigen, such as COS-7.
  • COS-7 cells to be used in transfection experiments were cultured in either 24 well cell culture plates (Corning) for cells to be used for protein extraction, or 4 chamber culture slides (Falcon) for cells to be used for staining.
  • the cells were grown in DMEM media supplemented with 10% FBS, but lacking penicillin/streptomycin, to 50 to 70% confluency.
  • Transfection medium sufficient for one well of a 24-well plate or culture slide was prepared by diluting 0.32 ⁇ g of plasmid DNA in 42.5 ⁇ l of serum-free DMEM and incubating the mixture at room temperature for 15 minutes.
  • LipofectAMINE (Gibco, BRL) was diluted in 42.5 ⁇ l of serum-free DMEM and incubated for 5 minutes at room temperature. After 5 minutes the LipofectAMINE mixture was added to the DNA mixture and incubated together at room temperature for 30 to 60 minutes. The cells to be transfected were washed once with serum-free DMEM before overlaying the transfection medium and the cells were placed back in the growth chamber for 4 hours.
  • DMEM +20% FBS 0.17 ml of DMEM +20% FBS was added to the cells.
  • the cells were the cultured for a further 40 hours before either being induced to undergo apoptosis prior to staining or harvested for Western blot analysis.
  • mock transfections were also performed in which the plasmid DNA was omitted from the transfection medium.
  • Protein was isolated for Western blotting from transfected cells by washing the cells twice in PBS (8 g/L NaCl, 0.2 g/L KCl, 1.44 g/L Na 2 HPO 4 , and 0.24 g/L KH 2 PO 4 ) and then adding 150 ⁇ l of hot SDS gel-loading buffer (50 mM Tris-HCl pH 6.8, 100 mM dithiothreitol, 2% SDS, 0.1% bromophenol blue, and 10% glycerol). The cell lysate was collected in a microcentrifuge tube, heated at 95° C. for 10 minutes, and then centrifuged at 13,000 ⁇ g for 10 minutes. The supernatant was transferred to a fresh microcentrifuge tube and stored at ⁇ 20° C. until ready for use.
  • PBS g/L NaCl, 0.2 g/L KCl, 1.44 g/L Na 2 HPO 4 , and 0.24 g/L KH 2 PO 4
  • the monoclonal antibody to p53 was used at a dilution of 0.1 ⁇ g/ml, and the monoclonal antibodies to Bcl-2 and c-Myc were both used at a dilution of 0.83 ⁇ g/ml. After incubation with primary antibody for 60 to 90 minutes, the membrane was washed 3 times for 15 minutes in PBS-T. Secondary antibody was then diluted in 1% milk in PBS and incubated with the membrane for 60 to 90 minutes. When p53 (Ab-6) was used as the primary antibody, the secondary antibody used was a goat anti-mouse IgG conjugated to alkaline phosphatase (Rockland) at a dilution of 1:1000.
  • Bcl-2 (Ab-1) and c-Myc (Ab-2) were used as the primary antibody
  • a rabbit anti-mouse IgG conjugated to peroxidase (Sigma) was used at a dilution of 1:5000. After incubation with the secondary antibody, the membrane was washed 3 times in PBS-T.
  • the colorimetric method was used only when p53 (Ab-6) was used as the primary antibody in conjunction with the alkaline phosphatase-conjugated secondary antibody. Bound antibody was visualized by incubating the blot in the dark in a solution of 0.33 mg/mL nitro blue tetrazolium, 0.165 mg/mL 5-bromo-4-chloro-3-indolyl phosphate, 100 mM NaCl, 5 mM MgCl 2 , and 100 mM Tris-HCl (pH 9.5).
  • the color reaction was stopped by incubating the blot in 2 mM EDTA in PBS.
  • a chemiluminescent detection method was used for all other primary antibodies, including anti-[HA]-peroxidase, Bcl-2 (Ab-1), and c-Myc (Ab-2).
  • the ECL Plus Western blotting detection kit (Amersham Pharmacia Biotech) was used to detect peroxidase-conjugated bound antibodies.
  • the membrane was lightly blotted dry and then incubated in the dark with a 40:1 mix of reagent A and reagent B for 5 minutes.
  • the membrane was blotted dry, placed between sheets of acetate, and exposed to X-ray film for time periods varying from 10 seconds to 10 minutes.
  • the percentage of apoptotic cells was determined 48 hours later by staining with either Hoescht or Annexin V-Cy3. Induction of apoptosis was also confirmed by Northern blot analyses, as shown in FIG. 22 .
  • the nuclear stain was used to label the nuclei of transfected COS-7 cells in order to identify apoptotic cells based on morphological features such as nuclear fragmentation and condensation.
  • a fixative consisting of a 3:1 mixture of absolute methanol and glacial acetic acid, was prepared immediately before use. An equal volume of fixative was added to the media of COS-7 cells growing on a culture slide and incubated for 2 minutes. The media/fixative mixture was removed from the cells and discarded, and 1 ml of fixative was added to the cells. After 5 minutes the fixative was discarded, and 1 ml of fresh fixative was added to the cells and incubated for 5 minutes.
  • the fixative was discarded, and the cells were air-dried for 4 minutes before adding 1 ml of Hoescht stain (0.5 ⁇ g/ml Hoescht 33258 in PBS). After a 10-minute incubation in the dark, the staining solution was discarded and the slide was washed 3 times for 1 minute with deionized water. After washing, 1 ml of Mcllvaine's buffer (0.021 M citric acid, 0.058 M Na 2 HPO 4 .7H 2 O; pH 5.6) was added to the cells, and they were incubated in the dark for 20 minutes. The buffer was discarded, the cells were air-dried for 5 minutes in the dark and the chambers separating the wells of the culture slide were removed.
  • Hoescht stain 0.5 ⁇ g/ml Hoescht 33258 in PBS. After a 10-minute incubation in the dark, the staining solution was discarded and the slide was washed 3 times for 1 minute with deionized water. After washing
  • Vectashield mounting media for fluorescence Vector Laboratories
  • Vector Laboratories A few drops of Vectashield mounting media for fluorescence (Vector Laboratories) was added to the slide and overlaid with a coverslip.
  • the stained cells were viewed under a fluorescence microscope using a UV filter. Cells with brightly stained or fragmented nuclei were scored as apoptotic.
  • An Annexin V-Cy3 apoptosis detection kit (Sigma) was used to fluorescently label externalized phosphatidylserine on apoptotic cells.
  • the kit was used according to the manufacturer's protocol with the following modifications.
  • transfected COS-7 cells growing on four chamber culture slides were washed twice with PBS and three times with 1 ⁇ Binding Buffer.
  • 150 ⁇ l of staining solution (1 ⁇ g/ml AnnCy3 in 1 ⁇ Binding Buffer) was added, and the cells were incubated in the dark for 10 minutes. The staining solution was then removed, and the cells were washed 5 times with 1 ⁇ Binding Buffer.
  • the chamber walls were removed from the culture slide, and several drops of 1 ⁇ Binding Buffer were placed on the cells and overlaid with a coverslip.
  • the stained cells were analyzed by fluorescence microscopy using a green filter to visualize the red fluorescence of positively stained (apoptotic) cells.
  • the total cell population was determined by counting the cell number under visible light.
  • the present example demonstrates modulation of apoptosis apoptosis-specific eIF-5A.
  • FIG. 23 is a flow chart illustrating the procedure for transient transfection of COS-7 cells, in which cells in serum-free medium were incubated in plasmid DNA in lipofectAMINE for 4 hours, serum was added, and the cells were incubated for a further 40 hours. The cells were then either incubated in regular medium containing serum for a further 48 hours before analysis (i.e. no further treatment), deprived of serum for 48 hours to induce apoptosis before analysis, or treated with actinomycin D for 48 hours to induce apoptosis before analysis.
  • FIG. 22 is a Western blot illustrating transient expression of foreign proteins in COS-7 cells following transfection with pHM6.
  • Protein was isolated from COS-7 cells 48 hours after either mock transfection, or transfection with pHM6-LacZ, pHM6-Antisense 3′ rF5A (pHM6-Antisense 3′ UTR rat apoptosis-specific eIF-5A), or pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A).
  • Five ⁇ g of protein from each sample was fractionated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with anti-[HA]-peroxidase. The bound antibody was detected by chemiluminescence and exposed to x-ray film for 30 seconds. Expression of LacZ (lane 2) and of sense rat apoptosis-specific eIF-5A (lane 4) is clearly visible.
  • COS-7 cells were either mock transfected or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A). Forty hours after transfection, the cells were induced to undergo apoptosis by withdrawal of serum for 48 hours.
  • the caspase proteolytic activity in the transfected cell extract was measured using a fluorometric homogenous caspase assay kit (Roche Diagnostics). DNA fragmentation was also measured using the FragEL DNA Fragmentation Apoptosis Detection kit (Oncogene) which labels the exposed 3′-OH ends of DNA fragments with fluorescein-labeled deoxynucleotides.
  • Additional COS-7 cells were either mock transfected or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A). Forty hours after transfection, the cells were either grown for an additional 48 hours in regular medium containing serum (no further treatment), induced to undergo apoptosis by withdrawal of serum for 48 hours or induced to undergo apoptosis by treatment with 0.5 ⁇ g/ml of Actinomycin D for 48 hours. The cells were either stained with Hoescht 33258, which depicts nuclear fragmentation accompanying apoptosis, or stained with Annexin V-Cy3, which depicts phosphatidylserine exposure accompanying apoptosis. Stained cells were also viewed by fluorescence microscopy using a green filter and counted to determine the percentage of cells undergoing apoptosis. The total cell population was counted under visible light.
  • FIG. 25 illustrates enhanced apoptosis as reflected by increased caspase activity when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specificeIF-5A in the sense orientation. Expression of rat apoptosis-specificeIF-5A resulted in a 60% increase in caspase activity.
  • FIG. 26 illustrates enhanced apoptosis as reflected by increased DNA fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specificeIF-5A in the sense orientation. Expression of rat apoptosis-specificeIF-5A resulted in a 273% increase in DNA fragmentation.
  • FIG. 27 illustrates detection of apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation. There is a greater incidence of fragmented nuclei in cells expressing rat apoptosis-specificeIF-5A.
  • FIG. 28 illustrates enhanced apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • Expression of rat apoptosis-specificeIF-5A resulted in a 27% and 63% increase in nuclear fragmentation over control in non-serum starved and serum starved samples, respectively.
  • FIG. 29 illustrates detection of apoptosis as reflected by phosphatidylserine exposure when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 30 illustrates enhanced apoptosis as reflected by increased phosphatidylserine exposure when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • Expression of rat apoptosis-specific eIF-5A resulted in a 140% and 198% increase in phosphatidylserine exposure over control, in non-serum starved and serum starved samples, respectively.
  • FIG. 31 illustrates enhanced apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specificeIF-5A in the sense orientation. Expression of rat apoptosis-specific eIF-5A resulted in a 115% and 62% increase in nuclear fragmentation over control in untreated and treated samples, respectively.
  • FIG. 32 illustrates a comparison of enhanced apoptosis under conditions in which COS-7 cells transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation were either given no further treatment or treatment to induce apoptosis.
  • the present example demonstrates modulation of apoptotic activity following administration of apoptosis-specific eIF-5A.
  • COS-7 cells were either mock transfected, transfected with pHM6-LacZ or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A) and incubated for 40 hours.
  • Five ⁇ g samples of protein extract from each sample were fractionated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with a monoclonal antibody that recognizes Bcl-2.
  • Rabbit anti-mouse IgG conjugated to peroxidase was used as a secondary antibody, and bound antibody was detected by chemiluminescence and exposure to x-ray film. Results are shown in FIG. 33 .
  • Bcl-2 is detectable in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ; thus showing that Bcl-2 is down-regulated with the pHM6-sense rF5A construct.
  • Additional COS-7 cells were either mock transfected, transfected with pHM6-antisense 3′ rF5A (pHM6-antisense 3′ UTR of rat apoptosis-specific eIF-5A) or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A). Forty hours after transfection, the cells were induced to undergo apoptosis by withdrawal of serum for 48 hours. Five ⁇ g samples of protein extract from each sample were fractionated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with a monoclonal antibody that recognizes Bcl-2. Rabbit anti-mouse IgG conjugated to peroxidase was used as a secondary antibody, and bound antibody was detected by chemiluminescence and exposure to x-ray film.
  • COS-7 cells were either mock transfected, transfected with pHM6-LacZ or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A) and incubated for 40 hours.
  • Five ⁇ g samples of protein extract from each sample were fractionated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with a monoclonal antibody that recognizes p53.
  • Goat anti-mouse IgG conjugated to alkaline phosphatase was used as a secondary antibody, and bound antibody was detected a colorimetrically.
  • COS-7 cells were either mock transfected, transfected with pHM6-LacZ or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A) and incubated for 40 hours.
  • Five ⁇ g samples of protein extract from each sample were fractionated by SDS-PAGE, transferred to a PVDF membrane, and probed with a monoclonal antibody that recognizes p53.
  • Corresponding protein blots were probed with anti-[HA]-peroxidase to determine the level of rat apoptosis-specific eIF-5A expression.
  • Goat anti-mouse IgG conjugated to alkaline phosphatase was used as a secondary antibody, and bound antibody was detected by chemiluminescence.
  • FIG. 33 illustrates downregulation of Bcl-2 when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • the upper panel illustrates the Coomassie-blue-stained protein blot; the lower panel illustrates the corresponding Western blot. Less Bcl-2 is detectable in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ.
  • FIG. 34 illustrates upregulation of Bcl-2 when COS-7 cells were transiently transfected with pHM6 containing the 3′ end of apoptosis-specific eIF-5A in the antisense orientation.
  • the upper panel illustrates the Coomassie-blue-stained protein blot; the lower panel illustrates the corresponding Western blot. More Bcl-2 is detectable in cells transfected with pHM6-antisense 3′ rF5A than in those mock transfected or transfected with pHM6-Sense rF5A.
  • FIG. 35 illustrates upregulation of c-Myc when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • the upper panel illustrates the Coomassie-blue-stained protein blot; the lower panel illustrates the corresponding Western blot. Higher levels of c-Myc is detected in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ or the mock control.
  • FIG. 36 illustrates upregulation of p53 when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • the upper panel illustrates the Coomassie-blue-stained protein blot; the lower panel illustrates the corresponding Western blot.
  • Higher levels of p53 is detected in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ or the mock control.
  • FIG. 37 illustrates the dependence of p53 upregulation upon the expression of pHM6-full length rat apoptosis-specificeIF-5A in COS-7 cells. More rat apoptosis-specificeIF-5A is detectable in the first transfection than in the second transfection.
  • the panel illustrates a corresponding Coomassie-blue-stained protein blot and the panel illustrates the Western blot with p53.
  • more p53 is detectable in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ or the mock control.
  • FIG. 47 depicts an experiment run on heart tissue to mimic the beating of a human heart and the subsequent induced heart attack.
  • FIG. 49 shows the laboratory bench set up. A slice of human heart tissue removed during valve replacement surgery was hooked up to electrodes. A small weight was attached to the heart tissue to ease in measuring the strength of the heart beats. The electrodes provided an electrical stimulus to get the tissue to start beating. The levels of gene expression for both apoptosis-specific eIF-5A and proliferating eIF-5A were measured in the heart tissue before ischemia was induced. See FIG. 46 . In the pre-ischemic heart tissue low levels both apoptosis-specific eIF-5A and proliferating eIF-5A were produced and their levels were in relative balance.
  • the heart did not beat as strong, as indicated by less compression/movement of the attached weight, thus indicating that the heart tissue cells were being killed rapidly due. to the presence of apoptosis-specific eIF-5A.
  • the EKG results are depicted in FIG. 48 .
  • a normal heart beat is shown (the pre-ischemic heart tissue).
  • the EKG shows decreased activity due to muscle cell death.
  • the EKG shows relative loss in strength of heart beat.
  • the following examples provide cell culture conditions.
  • Optic nerve heads (with attached pole) were removed and placed in Dulbecco's modified Eagle's medium (DMEM) supplemented with antibiotic/antimycotic, glutamine, and 10% FBS for 3 hours.
  • DMEM Dulbecco's modified Eagle's medium
  • the optic nerve head (ONH) button was retrieved from each tissue sample and minced with fine dissecting scissors into four small pieces.
  • Explants were cultured in 12.5 cm 2 plastic culture flasks in DMEM medium. Growth was observed within one month in viable explants. Once the cells reached 90% confluence, they were trypsinized and subjected to differential subculturing to produce lamina cribrosa (LC) and astrocyte cell populations.
  • LC lamina cribrosa
  • LC cells were subcultured in 25 cm 2 flasks in DMEM supplemented with gentamycin, glutamine, and 10% FBS, whereas astrocytes were expanded in 25 cm 2 flasks containing EBM complete medium (Clonetics) with no FBS. FBS was added to astrocyte cultures following 10 days of subculture. Cells were maintained and subcultured as per this protocol.
  • Cell populations obtained by differential subculturing were characterized for identity and population purity using differential fluorescent antibody staining on 8 well culture slides.
  • Cells were fixed in 10% formalin solution and washed three times with Dulbecco's Phosphate Buffered Saline (DPBS).
  • DPBS Dulbecco's Phosphate Buffered Saline
  • antibodies were diluted in 1% BSA in DPBS and applied to the cells in 6 of the wells. The remaining two wells were treated with only 1% bovine serum albumin (BSA) solution and no primary antibody as controls.
  • BSA bovine serum albumin
  • Cells were incubated with the primary antibodies for one hour at room temperature and then washed three times with DPBS.
  • Appropriate secondary antibodies were diluted in 1% BSA in DPBS, added to each well and incubated for 1 hour.
  • Immunofluorescent staining was viewed under a fluorescent microscope with appropriate filters and compared to the control wells that were not treated with primary antibody. All primary antibodies were obtained from Sigma unless otherwise stated. All secondary antibodies were purchased from Molecular Probes. Primary antibodies used to identify LC cells were: anti-collagen I, anti-collagen IV, anti-laminin, anti-cellular fibronectin. Primary antibodies used to identify astrocytes were: anti-galactocerebroside (Chemicon International), anti-A2B5 (Chemicon International), anti-NCAM, anti-human Von willebrand Factor. Additional antibodies used for both cell populations included anti-glial fibrillary (GFAP) and anti-alpha-smooth muscle actin.
  • GFAP anti-glial fibrillary
  • Cell populations were determined to be comprised of LC cells if they stained positively for collagen I, collagen IV, laminin, cellular fibronectin, alpha smooth muscle actin and negatively for glial fibrillary (GFAP).
  • Cell populations were determined to be comprised of astrocytes if they stained positively for NCAM, glial fibrillary (GFAP), and negatively for galactocerebroside, A2B5, human Von willebrand Factor, and alpha smooth muscle actin.
  • LC cell lines #506, #517, and #524 were established from the optic nerve heads of and 83-year old male, a 17-year old male, and a 26-year old female, respectively. All LC cell lines have been fully characterized and found to contain greater than 90% LC cells.
  • RKO American Type Culture Collection CRL-2577
  • a human colon carcinoma cell line expressing wild-type p53 was used to test the antisense oligonucleotides for the ability to suppress apoptosis-specific eIF-5A protein expression.
  • RKO were cultured in Minimum Essential Medium Eagle (MEM) with non-essential amino acids, Earle's salts, and L-glutamine. The culture media was supplemented with 10% fetal bovine serum (FBS) and 100 units of penicillin/streptomycin. The cells were grown at 37° C. in a humidified environment of 5% CO 2 and 95% air.
  • MEM Minimum Essential Medium Eagle
  • FBS fetal bovine serum
  • the cells were subcultured every 3 to 4 days by detaching the adherent cells with a solution of 0.25% trypsin and 1 mM EDTA.
  • the detached cells were dispensed at a split ratio of 1:10 to 1:12 into a new culture dish with fresh media.
  • HepG2 a human hepatocellular carcinoma cell line, was used to test the ability of an antisense oligo directed against human apoptosis-specific eIF-5A to block production of TNF- ⁇ in response to treatment with IL-1 ⁇ .
  • HepG2 cells were cultured in DMEM supplemented with gentamycin, glutamine, and 10% FBS and grown at 37° C. in a humidified environment of 5% CO 2 and 95% air.
  • Apoptosis was induced in RKO and lamina cribrosa cells using Actinomycin D, an RNA polymerase inhibitor, and camptothecin, a topoisomerase inhibitor, respectively. Actinomycin D was used at a concentration of 0.25 ⁇ g/ml and camptothecin was used at a concentration of 20, 40, or 50 ⁇ M. Apoptosis was also induced in lamina cribrosa cells using a combination of camptothecin (50 ⁇ M) and TNF- ⁇ (10 ng/ml). The combination of camptothecin and TNF- ⁇ was found to be more effective at inducing apoptosis than either camptothecin or TNF- ⁇ alone.
  • a set of three antisense oligonucleotides targeted against human apoptosis-specific eIF-5A were designed by, and purchased from, Molecula Research Labs.
  • the sequence of the first antisense oligonucleotide targeted against human apoptosis-specific eIF-5A (#1) was 5′ CCT GTC TCG AAG TCC AAG TC 3′ (SEQ ID NO: 63).
  • the sequence of the second antisense oligonucleotide targeted against human apoptosis-specific eIF-5A was 5′ GGA CCT TGG CGT GGC CGT GC 3′ (SEQ ID NO: 64).
  • the sequence of the third antisense oligonucleotide targeted against human apoptosis-specific eIF-5A was 5′ CTC GTA CCT CCC CGC TCT CC 3′ (SEQ ID NO: 65).
  • the control oligonucleotide had the sequence 5′ CGT ACC GGT ACG GTT CCA GG 3′ (SEQ ID NO: 66).
  • a fluorescein isothiocyanate (FITC)-labeled antisense oligonucleotide (Molecula Research Labs) was used to monitor transfection efficiency and had the sequence 5′ GGA CCT TGG CGT GGC CGT GCX 3′ (SEQ ID NO: 67), where X is the FITC label. All antisense oligonucleotides were fully phosphorothioated.
  • RKO cells were transfected with antisense oligonucleotides using the transfection reagent, Oligofectamine (Invitrogen). Twenty four hours prior to transfection, the cells were split onto a 24 well plate at 157,000 per well in MEM media supplemented with 10% FBS but lacking penicillin/streptomycin. Twenty four hours later the cells had generally reached a confluency of approximately 50%. RKO cells were either mock transfected, or transfected with 100 nM or 200 nM of antisense oligonucleotide.
  • Transfection medium sufficient for one well of a 24 well plate was prepared by diluting 0, 1.25, or 2.5 ⁇ l of a 20 ⁇ M stock of antisense oligonucleotide with serum-free MEM to a final volume of 42.5 ⁇ l and incubating the mixture at room temperature for 15 minutes.
  • 1.5 ⁇ l of Oligofectamine was diluted in 6 ⁇ l of serum-free MEM and incubated for 7.5 minutes at room temperature. After 5 minutes the diluted Oligofectamine mixture was added to the DNA mixture and incubated together at room temperature for 20 minutes.
  • the cells were washed once with serum-free MEM before adding 200 ⁇ l of MEM to the cells and overlaying 50 ⁇ l of transfection medium. The cells were placed back in the growth chamber for 4 hours.
  • Transfection of lamina cribrosa cells was also tested using 100 and 200 nM antisense oligonucleotide and Oligofectamine using the same procedure described for RKO cells.
  • effective transfection of lamina cribrosa cells was achieved by simply adding antisense oligonucleotide, diluted from 1 ⁇ M to 10 ⁇ M in serum-free media, to the cells for 24 hours and thereafter replacing the media with fresh antisense oligonucleotides diluted in serum-containing media every 24 hours for a total of two to five days.
  • antisense oligonucleotide transfection was optimized and monitored by performing transfections with an FITC-labeled antisense oligonucleotide having the same sequence as apoptosis-specific eIF-5A antisense oligonucleotide #2 (SEQ ID NO:64) but conjugated to FITC at the 3′ end.
  • RKO and lamina cribrosa cells were transfected with the FITC-labeled antisense oligonucleotide on an 8-well culture slide. Forty-eight hours later the cells were washed with PBS and fixed for 10 minutes in 3.7% formaldehyde in PBS.
  • the wells were removed and mounting media (Vectashield) was added, followed by a coverslip.
  • the cells were then visualized under UV light on a fluorescent microscope nucleus using a fluorescein filter (Green H546, filter set 48915) and cells fluorescing bright green were determined to have taken up the oligonucleotide.
  • the nuclear stain was used to label the nuclei of lamina cribosa cells in order to identify apoptotic cells based on morphological features such as nuclear fragmentation and condensation.
  • a fixative consisting of a 3:1 mixture of absolute methanol and glacial acetic acid, was prepared immediately before use. An equal volume of fixative was added to the media of cells growing on a culture slide and incubated for 2 minutes. The media/fixative mixture was removed from the cells and discarded and 1 ml of fixative was added to the cells. After 5 minutes the fixative was discarded and 1 ml of fresh fixative was added to the cells and incubated for 5 minutes.
  • the fixative was discarded and the cells were air-dried for 4 minutes before adding 1 ml of Hoescht stain (0.5 ⁇ g/ml Hoescht 33258 in PBS). After a 10 minute incubation in the dark, the staining solution was discarded, the chambers separating the wells of the culture slide were removed, and the slide was washed 3 times for 1 minute with deionized water. After washing, a few drops of Mcllvaine's buffer (0.021 M citric acid, 0.058 M Na 2 HPO 4 .7H 2 O; pH 5.6) was added to the cells and overlaid with a coverslip. The stained cells were viewed under a fluorescent microscope using a UV filter. Cells with brightly stained or fragmented nuclei were scored as apoptotic. A minimum of 200 cells were counted per well.
  • Hoescht stain 0.5 ⁇ g/ml Hoescht 33258 in PBS.
  • the DeadEndTM Fluorometric TUNEL (Promega) was used to detect the DNA fragmentation that is a characteristic feature of apoptotic cells. Following Hoescht staining, the culture slide was washed briefly with distilled water, and further washed by immersing the slide twice for 5 minutes in PBS (137 mM NaCl, 2.68 mM KCl, 1.47 mM KH 2 PO 4 , 8.1 mM Na 2 HPO 4 ), blotting the slide on paper towel between washes. The cells were permeabilized by immersing them in 0.2% Triton X-100 in PBS for 5 minutes.
  • the cells were then washed again by immersing the slide twice for 5 minutes in PBS and blotting the slide on paper towel between washes. 25 ⁇ l of equilibration buffer [200 mM potassium cacodylate (pH 6.6), 25 mM Tris-HCl (pH 6.6), 0.2 mM dithiothreitol, 0.25 mg/ml bovine serum albumin, and 2.5 mM cobalt chloride] was added per well and incubated for 5 to 10 minutes.
  • equilibration buffer 200 mM potassium cacodylate (pH 6.6), 25 mM Tris-HCl (pH 6.6), 0.2 mM dithiothreitol, 0.25 mg/ml bovine serum albumin, and 2.5 mM cobalt chloride
  • reaction mixture was prepared for each well by mixing in a ratio of 45:5:1, respectively, equilibration buffer, nucleotide mix [50 ⁇ M fluorescein-12-dUTP, 100 ⁇ M dATP, 10 mM Tris-HCl (pH 7.6), and 1 mM EDTA], and terminal deoxynucleotidyl transferase enzyme (Tdt, 25 U/ ⁇ l).
  • nucleotide mix 50 ⁇ M fluorescein-12-dUTP, 100 ⁇ M dATP, 10 mM Tris-HCl (pH 7.6), and 1 mM EDTA
  • Tdt terminal deoxynucleotidyl transferase enzyme
  • the reaction was terminated by immersing the slide in 2 ⁇ SSC [0.3 M NaCl, and 30 mM sodium citrate (pH 7.0)] and incubating for 15 minutes. The slide was then washed by immersion in PBS three times for 5 minutes. The PBS was removed by sponging around the wells with a Kim wipe, a drop of mounting media (Oncogene research project, JA1750-4ML) was added to each well, and the slide was overlayed with a coverslip. The cells were viewed under a fluorescent microscope using a UV filter (UV-G 365, filter set 487902) in order to count the Hoescht-stained nuclei. Any cells with brightly stained or fragmented nuclei were scored as apoptotic.
  • UV filter UV-G 365, filter set 487902
  • the cells were then viewed using a fluorescein filter (Green H546, filter set 48915) and any nuclei fluorescing bright green were scored as apoptotic.
  • the percentage of apoptotic cells in the field of view was calculated by dividing the number of bright green nuclei counted using the fluorescein filter by the total number of nuclei counted under the UV filter. A minimum of 200 cells were counted per well.
  • FIGS. 54-57 depict the results of these studies.
  • the percentage of apoptotic cells in samples having been transfected with apoptosis-specific eIF-5A is clearly much less than seen in cells having been transfected with the control oligonucleotide.
  • Protein from transfected RKO cells was harvested for Western blot analysis by washing the cells with PBS, adding 40 ⁇ l of hot lysis buffer [0.5% SDS, 1 mM dithiothreitol, 50 mM Tris-HCl (pH 8.0)] per well. The cells were scraped and the resulting extract was transferred to a microfuge tube, boiled for 5 minutes, and stored at—20° C. The protein was quantitated using the Bio-Rad Protein Assay (Bio-Rad) according to the manufacturer's instructions.
  • a monoclonal antibody from Oncogene which recognizes p53 (Ab-6) and a polyclonal antibody directed against a synthetic peptide (amino-CRLPEGDLGKEIEQKYD-carboxy) (SEQ ID NO:68) homologous to the c-terminal end of human apoptosis-specific eIF-5A that was raised in chickens (Gallus Immunotech).
  • An anti- ⁇ -actin antibody (Oncogene) was also used to demonstrate equal loading of protein.
  • the monoclonal antibody to p53 was used at a dilution of 0.05 ⁇ g/ml, the antibody against apoptosis-specific eIF-5A was used at a dilution of 1:1000, and the antibody against actin was used at a dilution of 1:20,000.
  • the membrane was washed 3 times for 15 minutes in 0.05% Tween-20/PBS. Secondary antibody was then diluted in 1% milk in 0.025% Tween-20/PBS and incubated with the membrane for 60 to 90 minutes.
  • the secondary antibody used was a rabbit anti-mouse IgG conjugated to peroxidase (Sigma) at a dilution of 1:5000.
  • a rabbit anti-chicken IgY conjugated to peroxidase (Gallus Immunotech) was used at a dilution of 1:5000.
  • the secondary antibody used with actin was a goat anti-mouse IgM conjugated to peroxidase (Calbiochem) used at a dilution of 1:5000. After incubation with the secondary antibody, the membrane was washed 3 times in PBS-T.
  • the ECL Plus Western blotting detection kit (Amersham Pharmacia Biotech) was used to detect peroxidase-conjugated bound antibodies.
  • the membrane was lightly blotted dry and then incubated in the dark with a 40:1 mix of reagent A and reagent B for 5 minutes.
  • the membrane was blotted dry, placed between sheets of acetate, and exposed to X-ray film for time periods varying from 10 seconds to 30 minutes.
  • the membrane was stripped by submerging the membrane in stripping buffer [100 mM 2-Mercaptoethanol, 2% SDS, and 62.5 mM Tris-HCl (pH 6.7)], and incubating at 50° C. for 30 minutes.
  • the membrane was then rinsed in deionized water and washed twice for 10 minutes in large volumes of 0.05% Tween-20/PBS. Membranes were stripped and re-blotted up to three times.
  • siRNAs Small inhibitory RNAs directed against human apoptosis-specific eIF-5A were used to specifically suppress expression of apoptosis-specific eIF-5A in RKO and lamina cribrosa cells.
  • Six siRNAs were generated by in vitro transcription using the SilencerTM siRNA Construction Kit (Ambion Inc.).
  • siRNAs #1 to #4 were generated against human apoptosis-specific eIF-5A (siRNAs #1 to #4)(SEQ ID NO:30-33).
  • siRNA #5 siRNA #5(SEQ ID NO: 34) which had the reverse sequence of the apoptosis-specific eIF-5A siRNA #1 (SEQ ID NO:30) but does not itself target apoptosis-specific eIF-5A.
  • the siRNAs were generated according to the manufacturer's protocol. In brief, DNA oligonucleotides encoding the desired siRNA strands were used as templates for T7 RNA polymerase to generate individual strands of the siRNA following annealing of a T7 promoter primer and a fill-in reaction with Klenow fragment.
  • the SilencerTM siRNA Labeling Kit—FAM (Ambion) was used to label GAPDH siRNA with FAM in order to monitor the uptake of siRNA into RKO and lamina cribrosa cells. After transfection on 8-well culture slides, cells were washed with PBS and fixed for 10 minutes in 3.7% formaldehyde in PBS. The wells were removed and mounting media (Vectashield) was added, followed by a coverslip. Uptake of the FAM-labeled siRNA was visualized under a fluorescent microscope under UV light using a fluorescein filter. The GAPDH siRNA was labeled according to the manufacturer's protocol.
  • RKO cells and lamina cribrosa cells were transfected with siRNA using the same transfection protocol.
  • RKO cells were seeded the day before transfection onto 8-well culture slides or 24-well plates at a density of 46,000 and 105,800 cells per well, respectively.
  • Lamina cribrosa cells were transfected when cell confluence was at 40 to 70% and were generally seeded onto 8-well culture slides at 7500 to 10,000 cells per well three days prior to transfection.
  • Transfection medium sufficient for one well of an 8-well culture slide was prepared by diluting 25.5 pmoles of siRNA stock to a final volume of 21.2 ⁇ l in Opti-Mem (Sigma).
  • Lipofectamine 2000 was diluted to a final volume of 21.2 ⁇ l in Opti-Mem and incubated for 7 to 10 minutes at room temperature. The diluted Lipofectamine 2000 mixture was then added to the diluted siRNA mixture and incubated together at room temperature for 20 to 30 minutes. The cells were washed once with serum-free media before adding 135 ⁇ l of serum-free media to the cells and overlaying the 42.4 ⁇ l of transfection medium. The cells were placed back in the growth chamber for 4 hours. After the incubation, 65 ⁇ l of serum-free media +30% FBS was added to the cells. Transfection of siRNA into cells to be used for Western blot analysis were performed in 24-well plates using the same conditions as the transfections in 8-well slides except that the volumes were increased by 2.3 fold.
  • lamina cribrosa cells were incubated for 72 hours prior to collection of cellular extract for Western blot analysis.
  • lamina cribrosa cells were treated with 50 ⁇ M of camptothecin (Sigma) and 10 ng/ml of TNF- ⁇ (Leinco Technologies) to induce apoptosis either 48 or 72 hours after transfection.
  • the cells were stained with Hoescht either 24 or 48 hours later in order to determine the percentage of cells undergoing apoptosis.
  • HepG2 cells were plated at 20,000 cells per well onto 48-well plates. Seventy two hours later the media was removed and fresh media containing either 2.5 ⁇ M control antisense oligonucleotide or 2.5 ⁇ M antisense oligonucleotide apoptosis-specific eIF-5A #2 was added to the cells. Fresh media containing antisense oligonucleotides was added after twenty four hours. After a total of 48 hours incubation with the oligonucleotides, the media was replaced with media containing interleukin 1 ⁇ (IL-1 ⁇ , 1000 pg/ml; Leinco Technologies) and incubated for 6 hours. The media was collected and frozen ( ⁇ 20° C.) for TNF- ⁇ quantification.
  • IL-1 ⁇ interleukin 1 ⁇
  • RKO cells were either left untransfected, mock transfected, or transfected with 200 nM of antisense oligonucleotides apoptosis-specific eIF-5A #1, #2, or #3 (SEQ ID NO: 25, 26, and 27). RKO cells were also transfected with 100 nM of antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26). Forty-eight hours after transfection, the cells were treated with 0.25 ⁇ g/ml Actinomycin D.
  • FIG. 52 which shows the levels of protein produced by RKO cells after being treated with antisense oligo 1, 2 and 3 (to apoptosis-specific eIF-5a)(SEQ ID NO:25, 26, and 27, respectively).
  • the RKO cells produced less apoptosis-specific eIF-5A as well as less p53 after having been transfected with the antisense apoptosis-specific eIF-5A nucleotides.
  • the lamina cribrosa cell line #506 was either (A) transfected with 100 nM of FITC-labeled antisense oligonucleotide using Oligofectamine transfection reagent or (B) transfected with 10 ⁇ M of naked FITC-labeled antisense oligonucleotide diluted directly in serum-free media. After 24 hours fresh media containing 10% FBS and fresh antisense oligonucleotide diluted to 10 ⁇ M was added to the cells. The cells, (A) and (B), were fixed after a total of 48 hours and visualized on a fluorescent microscope under UV light using a fluorescein filter. FIG. 53 shows uptake of the flourescently labeled antisense oligonucleotide.
  • the lamina cribrosa cell line #506 was transfected with 10 ⁇ M of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of 4 days. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with either 20 ⁇ M or 40 ⁇ M camptothecin for 48 hours. Antisense oligonucleotide and camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht and TUNEL. See FIG. 54 .
  • the lamina cribrosa cell line #506 was transfected with 10 ⁇ M of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26). Twenty-four hours later the media was changed and fresh antisense oligonucleotides were added. Forty-eight hours after beginning antisense oligonucleotide treatment, the antisense-oligonucleotides were removed and the cells were treated with 20 ⁇ M camptothecin for 3 days. The camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht and TUNEL. See FIG. 55 .
  • the lamina cribrosa cell line #517 was transfected with 1 ⁇ M of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of five days. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with 20 ⁇ M camptothecin for either 3 or 4 days. Antisense oligonucleotide and camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht and TUNEL. See FIG. 56 .
  • the lamina cribrosa cell line #517 was transfected with 2.5 ⁇ M of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of five days. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with 40 ⁇ M camptothecin for 3 days. Antisense oligonucleotide and camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 57 .
  • the lamina cribrosa cell line #517 was transfected with either 1 ⁇ M or 2.5 ⁇ M of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of five days. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with 40 ⁇ M camptothecin for 3 days. Antisense oligonucleotide and camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 58 .
  • the lamina cribrosa cell line #517 was left either untreated, or was treated with 10 ng/ml TNF- ⁇ , 50 ⁇ M camptothecin, or 10 ng/ml TNF- ⁇ and 50 ⁇ M camptothecin.
  • the percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 59 .
  • the lamina cribrosa cell lines #506 and #517 were transfected with either 2.5 ⁇ M or 5 ⁇ M of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of two days.
  • Fresh media containing antisense oligonucleotides was added after 24 hours.
  • Forty-eight hours after beginning antisense oligonucleotide treatment the cells were treated with 50 ⁇ M camptothecin and 10 ng/ml TNF- ⁇ for 2 days. The percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 60 .
  • the lamina cribrosa cell lines #506, #517, and #524 were transfected with 2.5 ⁇ M of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of two days.
  • Fresh media containing antisense oligonucleotides was added after 24 hours.
  • Forty-eight hours after beginning antisense oligonucleotide treatment the cells were treated with 50 ⁇ M camptothecin and 10 ng/ml TNF- ⁇ for 2 days. The percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 61 .
  • the following experiments show that cells transfected with siRNAs targeted against apoptosis-specific eIF-5A expressed less apoptosis-specific eIF-5A. The experiments also show that siRNAs targeted against apoptosis-specific eIF-5A were able to reduce apoptosis.
  • the lamina cribrosa cell line #517 was transfected with 100 nM of FAM-labeled siRNA using Lipofectamine 2000 transfection reagent either with serum (A) or without serum (B) during transfection.
  • the cells, (A) and (B), were fixed after a total of 24 hours and visualized on a fluorescent microscope under UV light using a fluorescein filter. See FIG. 62 .
  • RKO cells were transfected with 100 nM of siRNA either in the presence or absence of serum during the transfection.
  • Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO:34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-SA (siRNA #1 to #4)(SEQ ID NO:30-33).
  • the cell extract was harvested and 5 ⁇ g of protein from each sample was separated on an SDS-PAGE gel, transferred to a PVDF membrane, and Western blotted with an antibody against apoptosis-specific eIF-5A. After chemiluminescent detection, the membrane was stripped and re-probed with an antibody against bcl-2. After chemiluminescent detection, the membrane was stripped again and re-probed with an antibody against actin. See FIG. 63 .
  • lamina Cribrosa cell lines #506 and #517 were transfected with 100 nM of siRNA.
  • Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO:34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-5A (siRNA #1 to #4)(SEQ ID NO:30-33).
  • the cell extract was harvested and 5 ⁇ g of protein from each sample was separated on an SDS-PAGE gel, transferred to a PVDF membrane, and Western blotted with an antibody against apoptosis-specific eIF-5A. After chemiluminescent detection, the membrane was stripped and re-probed with an antibody against actin. See FIG. 64 .
  • the lamina cribrosa cell line #506 was transfected with 100 nm of siRNA.
  • Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO: 34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-5A (siRNA #1 to #4)(SEQ ID NO:30-33).
  • the media was replaced with media containing 50 , ⁇ M camptothecin and 10 ng/ml TNF- ⁇ .
  • the percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 65
  • the lamina cribrosa cell line #506 was transfected with 100 nm of siRNA.
  • Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO:34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-5A (siRNA #1 to #4)(SEQ ID NO:30-33).
  • the media was replaced with media containing 50 ⁇ M camptothecin and 10 ng/ml TNF- ⁇ .
  • the percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 66 .
  • the lamina cribrosa cell line #506 was either left untransfected or was transfected with 100 nm of siRNA.
  • Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO:34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-5A (siRNA #1 to #4)(SEQ ID NO:30-33).
  • the media was replaced with media containing 50 ⁇ M camptothecin and 10 ng/ml TNF- ⁇ . Fresh media was also added to the untransfected, untreated control cells. Forty-eight hours later, the percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 67 .
  • FIG. 68 Photographs of Hoescht-stained lamina cribrosa cell line #506 transfected with siRNA and treated with camptothecin and TNF- ⁇ from the experiment described in FIG. 67 and example 13 are provided in FIG. 68 .
  • This example shows that treating a human cell line with antisense oligonucleotides directed against apoptosis-specific eIF-5A causes the cells to produce less TNF- ⁇ .
  • HepG2 cells were treated with 2.5 ⁇ M of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 for a total of two days.
  • Fresh media containing antisense oligonucleotides was added after 24 hours. Additional cells were left untreated for two days. Forty-eight hours after the beginning of treatment, the cells were treated with IL-1 ⁇ (1000 pg/ml) in fresh media for 6 hours. At the end of the experiment, the media was collected and frozen ( ⁇ 20° C.) for TNF- ⁇ quantification. TNF- ⁇ released into the media was measured using ELISA assays purchased from Assay Designs Inc. See FIG. 69 . Cells that were transfected with antisense oligonucleotides of apoptosis-specific eIF-5A produced less TNF- ⁇ .
  • HT-29 cells human colon adenocarcinoma
  • siRNA against apoptosis-specific eIF-5A or with a control siRNA with the reverse sequence.
  • the siRNA used is as follows:
  • control siRNA used is as follows:
  • % G/C 39 5′ AAACACAUCCUCCUCAGGUCG 3′ (SEQ ID NO: 80)
  • FIGS. 74A and B are the results of the same experiment except the cells were at a higher density.
  • U-937 is a human monocyte cell line that grows in suspension and will become adherent and differentiate into macrophages upon stimulation with PMA (ATCC Number CRL-1593.2)(cells not obtained directly from ATCC).
  • Cells were maintained in RPMI 1640 media with 2 mM L-glutamine, 1.5 g/L sodium bicarbonate, 4.5 g/L glucose, 10 mM HEPES, 1.0 mM sodium pyruvate and 10% fetal bovine serum in a 37° C. CO 2 (5%) incubator.
  • Cells were split into fresh media (1:4 or 1:5 split ratio) twice a week and the cell density was always kept between 105 and 2 ⁇ 106 cells/ml.
  • Cells were cultured in suspension in tissue culture-treated plastic T-25 flasks and experiments were conducted in 24-well plates.
  • the cell density was adjusted to 3 ⁇ 105 cells/ml media.
  • the cells were harvested in log phase.
  • the cell suspension was transferred to 15 ml tubes and centrifuged at 400 ⁇ g for 10 mins at room temperature. The supernatant was aspirated and the cell pellet was washed/resuspended with fresh media. The cells were again centrifuged at 400 ⁇ g for 10 mins, the supernatant was aspirated, and the cell pellet was finally resuspended in fresh media.
  • Equal volumes of cell suspension and trypan blue solution (0.4% trypan blue dye in PBS) were mixed and the live cells were counted using a haemocytometer and a microscope. The cells were diluted to 4 ⁇ 105 cells/ml.
  • a 24-well plate was prepared by adding either PMA or DMSO (vehicle control) to each well. 1 ml of cell suspension was added to each well so that each well contained 400,000 cells, 0.1% DMSO ⁇ 162 nM PMA. The cells were maintained in a 37° C. CO 2 (5%) incubator. Separate wells of cells were harvested at times 0, 24, 48, 72, 96, 99 and 102 h. See FIG. 76 for a summary of the experimental time points and additions.
  • the media was changed at 72 h. Since some cells were adherent and others were in suspension, care was taken to avoid disrupting the adherent cells.
  • the media from each well was carefully transferred into corresponding microcentrifuge tubes and the tubes were centrifuged at 14,000 ⁇ g for 3 min. The tubes were aspirated, the cell pellets were resuspended in fresh media (1 ml, ( ⁇ ) DMSO, ( ⁇ ) PMA), and returned to their original wells. The cells become quiescent in this fresh media without PMA.
  • LPS 100 ng/ml
  • the suspension cells and media were transferred from each well into microcentrifuge tubes.
  • the cells were pelleted at 14,000 ⁇ g for 3 min.
  • the media (supernatant) was transferred to clean tubes and stored ( ⁇ 20° C.) for ELISA/cytokine analysis.
  • the cells remaining in the wells were washed with PBS (1 ml, 37° C.) and this PBS was also used to wash the cell pellets in the corresponding microcentrifuge tubes.
  • the cells were pelleted again at 14,000 ⁇ g for 3 min.
  • the cells were lysed with boiling lysis buffer (50 mM Tris pH 7.4 and 2% SDS).
  • the adherent cells and the suspension cells from each well were pooled.
  • the samples were boiled and then stored at ⁇ 20° C.
  • the protein concentration in each cell sample was determined by the BCA (bicinchoninic acid) method using BSA (bovine serum albumin) as the standard protein. Protein samples (5 ⁇ g total protein) were separated by 12% SDS-PAGE electrophoresis and transferred to PVDF membranes. The membranes were blocked with polyvinyl alcohol (1 ⁇ g/ml, 30 sec) and with 5% skim milk in PBS-t (1 h). The membranes were probed with a mouse monoclonal antibody raised against human eIF-5A (BD Biosciences cat #611976; 1:20,000 in 5% skim milk, 1 h). The membranes were washed 3 ⁇ 10 mins PBS-t.
  • the secondary antibody was a horseradish peroxidase-conjugated antimouse antibody (Sigma, 1:5000 in 1% skim milk, 1 h). The membranes were washed 3 ⁇ 10 mins PBS-t. The protein bands were visualized by chemiluminescence (ECL detection system, Amersham Pharmacia Biotech).
  • the membranes were stripped and reprobed for actin.
  • Membranes were stripped (100 mM 2-mercaptoethanol, 2% SDS, 62.5 mM Tris-HCl pH 6.7; 50° C. for 30 mins), washed, and then blocked as above.
  • actin primary antibody actin monoclonal antibody made in mouse; Oncogene, Ab-1; 1:20,000 in 5% skim milk.
  • the secondary antibody, washing, and detection were the same as above.
  • FIG. 77 shows that eIF5A is upregulated during monocyte (U-397) differentiation and subsequent TNF- ⁇ secretion.
  • HT-29 (human colon adenocarcinoma) cells were transfected with siRNA directed to apoptosis-specific eIF-5A. Approximately 48 hours after transfection the media was changed so that some of the test samples had media with interferon gamma and some of the samples had media without interferon gamma. 16 hours after interferon gamma addition, the cells were washed, and the media, with or without TNF-alpha, was placed on the cells. The media (used for ELISA detection of IL-8) and the cell lysate was harvested 8 or 24 hours later.
  • FIGS. 79 and 80 show that IL-8 is produced in response to TNF-alpha as well as in response to interferon. Priming the cells with interferon gamma prior to TNF treatment causes the cells to produce more IL-8 than either treatment alone. This may be due to the known upregulation of the TNF receptor 1 in response to interferon, so ‘priming’ the cells with interferon allows them to respond to TNF better since the cells have more receptors.
  • FIG. 81 is a western showing up-regulation (4 fold at 8 hours) of apoptosis-specific eIF-5A in response to interferon gamma in HT-29 cells.
  • LC cells were enriched by subculture in 25 cm 2 flasks in DMEM supplemented with gentamycin, glutamine, and 10% FBS. Cells were maintained and subcultured as per this protocol.
  • the identity and population purity of cells populations obtained by differential subculturing was characterized using differential fluorescent antibody staining on 8 well culture slides.
  • Cells were fixed in 10% formalin solution and washed three times with Dulbecco's Phosphate Buffered Saline (DPBS).
  • DPBS Dulbecco's Phosphate Buffered Saline
  • antibodies were diluted in 1% BSA in DPBS and applied to the cells in 6 of the wells.
  • the remaining two wells were treated with only 1% bovine serum albumin (BSA) solution and only secondary antibody as controls.
  • BSA bovine serum albumin
  • LC cells Appropriate secondary antibodies were diluted in 1% BSA in DPBS, added to each well and incubated for 1 hour. Following washing with DPBS, the slide was washed in water, air-dried, and overlayed with Fluoromount (Vector Laboratories). Immunofluorescent staining was viewed under a fluorescent microscope with appropriate filters and compared to the control wells that were not treated with primary antibody. All primary antibodies were obtained from Sigma unless otherwise stated. All secondary antibodies were purchased from Molecular Probes. Primary antibodies used to identify LC cells were: anti-collagen I, anti-collagen IV, anti-laminin, anti-cellular fibronectin, anti-glial fibrillary acidic protein (GFAP), and anti-alpha-smooth muscle actin.
  • GFAP anti-alpha-smooth muscle actin.
  • LC cell populations were determined to be comprised of LC cells if they stained positively for collagen I, collagen IV, laminin, cellular fibronectin, alpha smooth muscle actin and negatively for glial fibrillary (GFAP).
  • GFAP glial fibrillary
  • Apoptosis was induced in lamina cribrosa cells using a combination of 50 ⁇ M camptothecin (Sigma) and 10 ng/ml TNF- ⁇ (Leinco Technologies). The combination of camptothecin and TNF- ⁇ was found to be more effective at inducing apoptosis than either camptothecin or TNF- ⁇ alone.
  • siRNAs Small inhibitory RNAs directed against human apoptosis-specific eIF-5A were used to specifically suppress expression of eIF5A in lamina cribrosa cells.
  • Six siRNAs were generated by in vitro transcription using the SilencerTM siRNA Construction Kit (Ambion Inc.). Four siRNAs were generated against human apoptosis-specific eIF-5A (siRNAs #1 to #4). Two siRNAs were used as controls; an siRNA directed against GAPDH provided in the kit, and an siRNA (siRNA #5), which had the reverse sequence of the apoptosis-specific eIF-5A specific siRNA #1, but does not itself target eIF5A. The siRNAs were generated according to the manufacturer's protocol.
  • the eIF5A and control siRNA targets had the following sequences: siRNA # 1 5′ AAAGGAATGACTTCCAGCTGA 3′; (SEQ ID NO: 81) siRNA # 2 5′ AAGATCGTCGAGATGTCTACT 3′; (SEQ ID NO: 82) siRNA # 3 5′ AAGGTCCATCTGGTTGGTATT 3′; (SEQ ID NO: 83) siRNA # 4 5′ AAGCTGGACTCCTCCTACACA 3′; (SEQ ID NO: 84) siRNA # 5′ AAAGTCGACCTTCAGTAAGGA 3′. (SEQ ID NO: 85) Lamina cribrosa cells were transfected with siRNA using LipofectAMINE 2000.
  • Lamina cribrosa cells were transfected when cell confluence was at 40 to 70% and were generally seeded onto 8-well culture slides at 7500 cells per well three days prior to transfection.
  • Transfection medium sufficient for one well of an 8-well culture slide was prepared by diluting 25.5 pmoles of siRNA to a final volume of 21.2 ⁇ l in Opti-Mem (Sigma).
  • 0.425 ⁇ l of Lipofectamine 2000 was diluted to a final volume of 21.2 ⁇ l in Opti-Mem and incubated for 7 to 10 minutes at room temperature.
  • the diluted Lipofectamine 2000 mixture was then added to the diluted siRNA mixture and incubated together at room temperature for 20 to 30 minutes.
  • the cells were washed once with serum-free media before adding 135 ⁇ l of serum-free media to the cells and overlaying 42.4 ⁇ l of transfection medium. The cells were placed back in the growth chamber for 4 hours. After the incubation, 65 ⁇ l of serum-free media plus 30% FBS was added to the cells.
  • Transfection of siRNA into cells to be used for Western blot analysis were performed in 24-well plates using the same conditions as the transfections in 8-well slides except that the volumes were increased by 2.3 fold. Following transfection, lamina cribrosa cells were incubated for 72 hours prior to treatment with 50 ⁇ M of camptothecin (Sigma) and 10 ng/ml of TNF- ⁇ (Leinco Technologies) to induce apoptosis. Cell lysates were then harvested for Western blotting or the cells were examined for apoptosis
  • Transfected cells that had been treated with TNF- ⁇ and camptothecin for 24 hours were stained with Hoescht 33258 in order to determine the percentage of cells undergoing apoptosis. Briefly, cells were fixed with a 3:1 mixture of absolute methanol and glacial acetic acid and then incubated with Hoescht stain (0.5 pg/ml Hoescht 33258 in PBS). After a 10 minute incubation in the dark, the staining solution was discarded, the chambers separating the wells of the culture slide were removed, and the slide was washed 3 times for 1 minute with deionized water.
  • Hoescht stain 0.5 pg/ml Hoescht 33258 in PBS
  • Mcllvaine's buffer 0.021 M citric acid, 0.058 M Na 2 HPO 4 .7H 2 O; pH 5.6
  • the stained cells were viewed under a fluorescent microscope using a UV filter. Cells with brightly stained or fragmented nuclei were scored as apoptotic. A minimum of 200 cells were counted per well.
  • the DeadEndTM Fluorometric TUNEL (Promega) was also used to detect the DNA fragmentation that is a characteristic feature of apoptotic cells.
  • the culture slide was washed briefly with distilled water, and further washed by immersing the slide twice for 5 minutes in PBS (137 mM NaCl, 2.68 mM KCl, 1.47 mM KH 2 PO 4 , 8.1 mM Na 2 HPO 4 ), blotting the slide on paper towel between washes.
  • PBS 137 mM NaCl, 2.68 mM KCl, 1.47 mM KH 2 PO 4 , 8.1 mM Na 2 HPO 4
  • the cells were permeabilized by immersing them in 0.2% Triton X-100 in PBS for 5 minutes.
  • the cells were then washed again by immersing the slide twice for 5 minutes in PBS and blotting the slide on paper towel between washes.
  • equilibration buffer 200 mM potassium cacodylate (pH 6.6), 25 mM Tris-HCl (pH 6.6), 0.2 mM dithiothreitol, 0.25 mg/ml bovine serum albumin, and 2.5 mM cobalt chloride] was added per well and incubated for 5 to 10 minutes.
  • reaction mixture was prepared for each well by mixing in a ratio of 45:5:1, respectively, equilibration buffer, nucleotide mix [50 ⁇ M fluorescein-12-dUTP, 100 ⁇ M dATP, 10 mM Tris-HCl (pH 7.6), and 1 mM EDTA], and terminal deoxynucleotidyl transferase enzyme (Tdt, 25 U/ ⁇ l).
  • nucleotide mix 50 ⁇ M fluorescein-12-dUTP, 100 ⁇ M dATP, 10 mM Tris-HCl (pH 7.6), and 1 mM EDTA
  • Tdt terminal deoxynucleotidyl transferase enzyme
  • the reaction was terminated by immersing the slide in 2 ⁇ SSC [0.3 M NaCl, and 30 mM sodium citrate (pH 7.0)] and incubating for 15 minutes. The slide was then washed by immersion in PBS three times for 5 minutes. The PBS was removed by sponging around the wells with a Kim wipe, a drop of mounting media (Oncogene research project, JA1750-4ML) was added to each well, and the slide was overlayed with a coverslip. The cells were viewed under a fluorescent microscope using a UV filter (UV-G 365, filter set 487902) in order to count the Hoescht-stained nuclei. Any cells with brightly stained or fragmented nuclei were scored as apoptotic.
  • UV filter UV-G 365, filter set 487902
  • the cells were then viewed using a fluorescein filter (Green H546, filter set 48915) and any nuclei fluorescing bright green were scored as apoptotic.
  • the percentage of apoptotic cells in the field of view was calculated by dividing the number of bright green nuclei counted using the fluorescein filter by the total number of nuclei counted under the UV filter. A minimum of 200 cells were counted per well.
  • Protein was isolated for Western blotting from lamina cribrosa cells growing on 24-well plates by washing the cells twice in PBS (8 g/L NaCl, 0.2 g/L KCl, 1.44 g/L Na 2 HPO 4 , and 0.24 g/L KH 2 PO 4 ) and then adding 50 ⁇ l of lysis buffer [2% SDS, 50 mM Tris-HCl (pH 7.4)]. The cell lysate was collected in a microcentrifuge tube, boiled for 5 minutes and stored at ⁇ 20° C. until ready for use. Protein concentrations were determined using the Bicinchoninic Acid Kit (BCA; Sigma).
  • the primary antibody was preincubated for 30 minutes in a solution of 5% milk in PBS prior to incubation with the membrane.
  • the primary antibodies used were anti-eIF5A (BD Transduction Laboratories) at 1:20,000 and anti- ⁇ -actin (Oncogene).
  • the membranes were washed three times in PBS-T and incubated for 1 hour with the appropriate HRP-conjugated secondary antibodies diluted in 1% milk in PBS.
  • the blot was washed and the ECL Plus Western blotting detection kit (Amersham Pharmacia Biotech) was used to detect the peroxidase-conjugated bound antibodies.
  • LC cell lines Two lamina cribrosa (LC) cell lines were established from optic nerve heads obtained from male donors ranging in age from 83 years (#506) to 17 years (#517).
  • the cells isolated from the human lamina cribrosa had the same broad, flat morphology with prominent nucleus observed in other studies (Lambert et al., 2001). Consistent with the characterizations of other groups, the LC cells showed immunoreactivity to alpha smooth muscle actin ( FIG. 82 a ) as well as to a number of extracellular matrix proteins including cellular fibronectin ( FIG. 82 b ), laminin ( FIG.
  • TNF- ⁇ is believed to play an important role during the glaucomatous process
  • the susceptibility of LC cells to the cytotoxic effects of TNF- ⁇ was examined.
  • Confluent LC cells were exposed to either camptothecin, TNF- ⁇ , or a combination of camptothecin and TNF- ⁇ for 48 hours ( FIG. 83 ).
  • Hoescht staining revealed that TNF- ⁇ alone was not cytotoxic to LC cells.
  • Treatment with camptothecin resulted in approximately 30% cell death of the LC cells.
  • EIF5A is a nucleocytoplasmic shuttle protein known to be necessary for cell division and recently suggested to also be involved during apoptosis.
  • the expression of apoptosis-specific eIF-5A protein in LC cells being induced to undergo apoptosis by either camptothecin, or camptothecin plus TNF- ⁇ .
  • the expression of apoptosis-specific eIF-5A did not alter significantly upon treatment with camptothecin except perhaps to decrease slightly ( FIG. 84A ). However, a significant upregulation of apoptosis-specific eIF-5A protein was observed after 8 and 24 hours of camptothecin plus TNF- ⁇ treatment ( FIG. 84B ).
  • siRNAs #1 to #4 siRNAs #1 to #4 targeting apoptosis-specific eIF-5A were designed and synthesized by in vitro transcription.
  • LC cell lines #506 and #517 were transfected with each of the siRNAs and expression of apoptosis-specific eIF-5A protein in the cell lysate was examined 72 hours later ( FIG. 85 ).
  • siRNA #5 an siRNA against GAPDH and/or a control siRNA having the same chemical composition as siRNA #1 but which does not recognize apoptosis-specific eIF-5A. All siRNAs directed against apoptosis-specific eIF-5A were capable of significantly suppressing apoptosis-specific eIF-5A expression in both LC cell lines ( FIG. 85 ).
  • the GAPDH siRNA was used as an additional control because, unlike the control siRNA #5 which simply has the reverse sequence of siRNA #1 and does not have a cellular target, it is an active siRNA capable of suppressing the expression of it's target protein, GAPDH (data not shown).
  • siRNAs against apoptosis-specific eIF-5A were also capable of protecting transfected LC cells (#506) from apoptosis induced by 24 hour treatment with TNF- ⁇ and camptothecin ( FIG. 86 ).
  • siRNAs #1 to #4 were found to be able to reduce apoptosis of LC cells by 59% (siRNA #1), 35% (siRNA #2), 50% (siRNA #3), and 69% (siRNA #4).
  • siRNA against GAPDH was also able to reduce apoptosis of LC cells by 42% ( FIG. 86 ).
  • GAPDH is known to have cellular functions outside of it's role as a glycolytic enzyme, including a proposed function during apoptosis of cerebellar neurons (Ishitani and Chuang, 1996; Ishitani et al., 1996a; Ishitani et al., 1996b).
  • siRNA #1 was able to reduce apoptosis of the LC line #517 by 53% in response to TNF- ⁇ and camptothecin indicating that apoptosis-specific eIF-5A siRNAs are protective for LC cells isolated from different optic nerve heads ( FIG. 87 ).
  • LC cells exposed to TNF-a and camptothecin were dying by classical apoptosis
  • DNA fragmentation was evaluated in situ using the terminal deoxynucleotidyl transferase-mediated dUTP-digoxigenin nick end labeling (TUNEL) method.
  • LC cells (#506) were treated with TNF- ⁇ and camptothecin for 24 hours, 3 days after transfection with either an apoptosis-specific eIF-5A siRNA (siRNA #1) or a control siRNA (siRNA #5). The cells were also stained with Hoescht to facilitate visualization of the nuclei.
  • a 60% SIP (9 parts v/v Percoll with 1 part v/v 1.5M NaCl) was cushioned on the bottom of 15 ml conical tubes.
  • the blood was then layered overtop with minimal mixing of the blood and Percoll cushion.
  • the samples were centrifuged for 30 minutes total at 100 ⁇ g with slow acceleration in the first 5 minutes and slow deceleration in the last 5 minutes.
  • the pure serum at the very top of the resulting gradient was removed and the white cushion (1-2 ml) of PBMCs was collected and added dropwise to a tube containing 10 ml of warm RPMI plus 15% FBS.
  • the PBMCs were pelleted and counted.
  • PBMCs peripheral blood mononuclear cells
  • PBMCs peripheral blood mononuclear cells
  • the cells were treated with phorbol 12-myristate 13-acetate (PMA; 100 ng/well). At 72 hours the media was replaced and did not contain any stimulating factors. Then at 96 hours after PMA addition to PBMCs, lipopolysaccharide (LPS; 100 ng/well; from E. coli, serotype 0111) was added to the wells. Samples were collected before LPS addition (96 h), and at various times after addition as outlined in FIG. 91 . Both adherent cells (likely to be monocytes and macrophages) were collected with the floating cells (likely to be lymphocytes).
  • PMA phorbol 12-myristate 13-acetate
  • PBMCs were collected and seeded at 2 ⁇ 10 5 to 5 ⁇ 10 5 cells/well.
  • the PBMCs were stimulated with phytohemagglutinin (PHA; 100 ng/ml), phorbol 12-myristate 13-acetate (PMA; 100 ng/ml), lipopolysaccharide (LPS; 100 ng/ml) or all three (each at 100 ng/ml).
  • PHA phytohemagglutinin
  • PMA phorbol 12-myristate 13-acetate
  • LPS lipopolysaccharide
  • the samples were collected 12 and 36 hours after stimulation and analyzed for apoptosis-specific eIF-5A expression ( FIG. 94 ).
  • PBMCs were transfected the day they were prepared. Cells were seeded at 2 ⁇ 10 5 to 5 ⁇ 10 5 cells/well (150 ⁇ l per well for transfection in a 24-well plate). They were either transfected individually in each well (Donors 77, 78 and 79; FIGS. 95 and 96 ) or all at once in a conical tube before seeding (Donors 80 and 84; FIG. 96 ). For each well of cells to be transfected, 15 pmoles of siRNA was diluted in 50 ⁇ l of Opti-MEM (Sigma).
  • Lipofectamine 2000 (Invitrogen) was diluted in 49 ⁇ l of Opti-MEM, incubated for 7 to 10 minutes, added to the diluted siRNA and incubated 25 minutes.
  • the transfection medium was overlayed onto the cells were placed in the 37° C. growth chamber for 4 hours. The final transfection medium contained 9% serum. After the incubation, 250 ⁇ l of serum-free RPMI+21% FBS was added to the cells to make the final serum concentration 15%.
  • LPS lipopolysaccharide
  • the cells were washed with 1 ml of 37° C. phosphate buffered saline (PBS) and then lysed in boiling lysis buffer (50 mM Tris pH 7, 2% SDS; 100 ⁇ l per well). The cell lysates were boiled and stored frozen at ⁇ 20° C. for BCA protein quantitation.
  • PBS phosphate buffered saline
  • HT-29 a human colorectal adenocarcinoma cell line, was maintained in RPMI with 10% fetal bovine serum (FBS).
  • FBS fetal bovine serum
  • U937 a histiocytic lymphoma cell line, was grown in suspension in RPMI with 10% FBS. Both cell lines were maintained in a humidified environment at 37° C. and 5% CO 2 . For experiments with U937 cells, cells were counted and adjusted to 3 ⁇ 10 5 cells/ml two days before the start of the experiment.
  • siRNA sequences were designed based on the human apoptosis-specific eIF-5A sequence and were synthesized by Dharmacon RNA Technologies.
  • the apoptosis-specific eIF-5A siRNA (h5A1) target sequence was: 5′ NNGCUGGACUCCUCCUACACA 3′.
  • the corresponding double stranded siRNA sequence was: 5′ GCUGGACUCCUCCUACACAdTdT 3′ 3′ dTdTCGACCUGAGGAGGAUGUGU 5′
  • control siRNA (hcontrol) sequence was 5′ NNACACAUCCUCCUCAGGUCG 3′.
  • the corresponding double stranded siRNA sequence was: 5′ ACACAUCCUCCUCAGGUCGdTdT 3′ 3′ dTdTUGUGUAGGAGGAGUCCAGC 5′ Transfection of HT-29 Cells
  • HT-29 cells were seeded at 105,000 cells per well onto a 24-well plate.
  • 25.5 pmoles of siRNA was diluted in 50 ⁇ l of Opti-Mem (Sigma).
  • 1 ⁇ l of Lipofectamine 2000 (Invitrogen) was diluted in 49 ⁇ l of Opti-Mem, incubated for 7 to 10 minutes and added to the diluted siRNA and incubated 25 minutes.
  • the cells to be transfected were washed once with serum-free RPMI before adding 300 ⁇ l of serum-free RPMI and overlaying 100 ⁇ l of transfection medium. The cells were placed back in the growth chamber for 4 hours. After the incubation, 300 ⁇ l of serum-free RPMI+30% FBS was added to the cells.
  • apoptosis-specific eIF-5A and control siRNA were diluted in Opti-Mem media (Sigma). 400 ⁇ l cells (800,000 cells) and 100 pmoles siRNA were mixed in a 0.4 mm electroporation cuvette. The cells were electroporated at 300 V, 10 mSec, 1 pulse with an ECM 830 Electrosquare porator (BTX, San Diego, Calif.). Following electroporation, the cells were gently mixed and added to wells containing RPMI and concentrated FBS so that the final FBS concentration was 10%.
  • TNF- ⁇ production was induced in HT-29 cells according to the method developed by Suzuki et al. 2003.
  • HT-29 cells were primed with 200 units/ml interferon gamma (Roche Diagnostics) 48 hours after transfection. After 16 hours of interferon gamma (IFN ⁇ ) priming the cells were washed with media and lipopolysaccharide (LPS; 100 ng/ml; from E. coli, serotype 0111; Sigma) was added at 100 ⁇ g/ml. After 8 or 24 hours of LPS stimulation, the media from each well was transferred to microcentrifuge tubes and stored at ⁇ 20° C. until assayed for TNF ⁇ by ELISA.
  • LPS lipopolysaccharide
  • the cells were washed with 1 ml of phosphate buffered saline (PBS) heated to 37° C. and then lysed in boiling lysis buffer (50 mM Tris pH 7, 2% SDS). The cell lysates were boiled and stored frozen at ⁇ 20° C. The protein concentration in the cell lysates was determined by bicinchoninic acid assays (BCA) with bovine serum albumin used as the standard.
  • PBS phosphate buffered saline
  • BCA bicinchoninic acid assays
  • IL-8 production was induced in HT-29 cells by treatment with IFN ⁇ .
  • HT-29 cells were treated with 200 units/ml IFN ⁇ 48 hours after transfection. After 24 hours of treatment, the media from each well was transferred to microcentrifuge tubes and stored at ⁇ 20° C. until assayed for IL-8 by liquid-phase electrochemiluminescence (ECL).
  • ECL liquid-phase electrochemiluminescence
  • the cells were washed with 1 ml of phosphate buffered saline (PBS) heated to 37° C. and then lysed in boiling lysis buffer (50 mM Tris pH 7, 2% SDS).
  • the cell lysates were boiled and stored frozen at ⁇ 20° C.
  • the protein concentration in the cell lysates was determined by bicinchoninic acid assays (BCA) with bovine serum albumin used as the standard.
  • BCA bicinchoninic acid assays
  • U937 cells were collected and counted 16 hours after electroporation. 200,000 cells in 1 ml of media were added to each well of 24-well plates. Macrophage differentiation was stimulated by adding phorbol 12-myristate 13-acetate (PMA; 100 ng/ml). After 48 h with PMA, >80% of the monocytes had transformed from cells in suspension (monocytes) to adherent cells (macrophages). At 48 hours the media and any non-adherent cells were removed and fresh RPMI media with 10% FBS (1 ml per well) was added. The cells were left for 24 hours in fresh media to become quiescent.
  • PMA phorbol 12-myristate 13-acetate
  • LPS lipopolysaccharide
  • IFN ⁇ interferon ⁇
  • a combination of LPS and IFN ⁇ were added to the wells.
  • Samples were collected before stimulator addition (72 h), and at various times after addition as outlined in FIG. 108 .
  • the media from each well was transferred to clean microcentrifuge tubes and cleared of any debris by centrifugation at 13000 ⁇ g for 3 mins. The media was stored at ⁇ 20° C. in 200-250 ul aliquots prior to analysis.
  • the cells were washed with 1 ml of 37° C. phosphate buffered saline (PBS) and then lysed in boiling lysis buffer (50 mM Tris pH 7, 2% SDS; 75 ⁇ l per well). Like wells were pooled. The cell lysates were boiled and stored frozen at ⁇ 20° C.
  • PBS phosphate buffered saline
  • TNF ⁇ was quantified using ELISA kits from Assay Designs according to the manufacturer's instructions with supplied standards for 0-250 pg TNF ⁇ /ml.
  • media samples for TNF ⁇ were diluted 20 fold (0 h, 3 h LPS) or 80 fold (6 h, 24 h, 30 h LPS) with RPMI+10% FBS.
  • IL-1 ⁇ , IL-8, and IL-6 were quantified by liquid-phase electrochemiluminescence (ECL).
  • ECL liquid-phase electrochemiluminescence
  • IL-8, IL-1 ⁇ , and IL-6 were assayed by liquid-phase. Briefly, a purified monoclonal mouse anti- mouse anti-human IL-8, IL-6 or IL-1 ⁇ (R & D Systems) were labeled with biotin (Igen, Inc., Gaithersburg, Md.). In addition, the goat anti-human IL-8, IL-6, or IL-1 ⁇ antibody (R & D) were labeled with ruthenium (Igen) according to the manufacturer's instructions. The biotinylated antibodies were diluted to a final concentration of 1 mg/mL in PBS, pH 7.4, containing 0.25% BSA, 0.5% Tween-20 and 0.01% azide, (ECL buffer).
  • the protein concentration in the cell lysates was determined by bicinchoninic acid assays (BCA) with bovine serum albumin used as the standard. 5 ⁇ g of total cellular protein was separated by either 10% or 14% SDS-PAGE (sodium dodecyl sulfate polyacrylamide gel electrophoresis). 10% gels were used for analysis of proteins above 50 kDa (TLR4, IFN ⁇ , TNF-R1, iNOS) while 14% gels were used for apoptosis-specific eIF-5A (17 kDa).
  • PVDF polyvinylidene fluoride
  • eIF-5A BD Biosciences; 1:20,000; incubate 1 hour; recognizes both apoptosis-specific eIF-5A and eIF5A2
  • TLR4 Santa Cruz Biotechnology Inc; TLR4 (H-80): sc-10741; 1:1000; incubate 2 hours
  • IFN- ⁇ R ⁇ Santa Cruz Biotechnology Inc; IFN- ⁇ R ⁇ (C-20): sc-700; 1:1000; incubate 1 hour
  • TNF-R1 Santa Cruz Biotechnology Inc; TNF-R1 (H-5): sc-8436; 1:200; incubate 3 hours
  • iNOS BD Transduction Laboratories: 610431; 1:10,000; incubate 1 hour
  • ⁇ -actin Oncogene; actin (Ab-1); 1:20,000; incubate 1 hour).
  • HRP horseradish peroxidase-conjugated
  • Blots were washed 4 times for 5-10 mins with PBS-t. Blots were developed with enhanced chemiluminescent detection reagent (ECL; Amersham Pharmacia Biotech) according to the manufacturers instructions and bands were visualized on X-ray film (Fuji).
  • ECL enhanced chemiluminescent detection reagent
  • RT-PCR was performed according to Medvedev et al. 2002 in order to observe changes in TLR4 mRNA expression in transfected HT-29 cells in response to IFN ⁇ . Expression of GAPDH was used as a control to show that equal amounts of cDNA were being used between samples. Increasing PCR cycles (20, 25, 30, and 35) were used to determine the optimal cycle number that resulted in detectable amplified products under nonsaturating conditions. PCR products were detected by ethidium bromide-incorporation and were separated by agarose gel electrophoresis. RT-PCR of total MRNA isolated from siRNA-transfected HT-29 cells treated with or without IFN ⁇ for 6 hours was used to detect TLR4 and GAPDH transcripts.
  • HT-29 cells were transfected with siRNA as described above. 48 hours after transfection, the cells were treated with 200 units/ml IFN ⁇ . Control cells which were not treated with IFN ⁇ received only a media change. Total mRNA was isolated using the GenElute Mammalian RNA miniprep kit (Sigma) according to the manufacturer's protocol for adherent cells. The media was removed and the cells were washed twice with warm PBS. Lysis buffer was added to the cells and the lysate was transferred to a microcentrifuge tube and total RNA was isolated according to the manufacturer's protocol.
  • the primers for TLR4 were: Forward 5′ CGGATGGCAACATTTAGAATTAGT 3′ Reverse 5′ TGATTGAGACTGTAATCAAGAACC 3′ Expected fragment size: 674 bp
  • the primers for GAPDH were: Forward 5′ CTGATGCCCCCATGTTCGTCAT 3′ Reverse 5′ CCACCACCCTGTTGCTGTAG 3′ Expected fragment size: 599 bp
  • RNA reverse transcribed using the following conditions: Mix: RNA 2.5 ⁇ g Poly (T) primer 6.25 ⁇ l Depc water to 13.75 ⁇ l Heat 70° C. 5 min Chill on ice 5 min Add: 5 ⁇ AMV Buffer 5.0 ⁇ l dNTPs (10 mM) 2.5 ⁇ l Rnase Inhibitor 1.25 ⁇ l AMV RT 2.5 ⁇ l Heat 42° C. 60 min Heat 70° C. 10 min
  • a single PCR reaction was performed using the following conditions: 10 ⁇ Tsg buffer 2.0 ⁇ l dNTP (10 mM) 0.4 ⁇ l forward primer (25 pmol/ ⁇ l) 0.4 ⁇ l reverse primer (25 pmol/ ⁇ l) 0.4 ⁇ l MgCl 2 (15 mM) 2.0 ⁇ l cDNA 0.8 ⁇ l H 2 O 13.88 ⁇ l Tsg polymerase 0.12 ⁇ l
  • the PCR conditions for TLR4 were: Heat to 95° C. 5 min 20, 25, 30, or 35 cycles of: 95° C. 1 min 55° C. 1 min 72° C. 2 min Extend at 72° C. for 10 min Sink to 4° C.
  • the PCR conditions for GAPDH were: Heat to 95° C. 5 min 20, 25, 30, or 35 cycles of: 95° C. 1 min 57° C. 1 min 72° C. 2 min Extend at 72° C. for 10 min Sink to 4° C.

Abstract

The present invention relates to apoptosis specific eucaryotic initiation factor 5A (eIF-5A), referred to as apoptosis-specific eIF-5A or eIF5A1, nucleic acids and polypeptides and methods for inhibiting or suppressing apoptosis in cells using antisense nucleotides or siRNAs to inhibit expression of apoptosis-specific eIF-5A. The invention also relates to suppressing or inhibiting expression of pro-inflammatory cytokines or inhibiting activation of NFKB by inhibiting expression of apoptosis-specific eIF-5A.

Description

    RELATED APPLICATIONS
  • This application is a continuation-in-part of U.S. application Ser. No. 10/383,614, filed on Mar. 10, 2003, which is a continuation-in-part of Ser. No. 10/277,969, filed Oct. 23, 2002, which is a continuation-in-part of Ser. No. 10/200,148, filed on Jul. 23, 2002, which is a continuation-in-part of U.S. application Ser. No. 10/141,647, filed May 7, 2002, which is a continuation-in part of U.S. application Ser. No. 9/909,796, filed Jul. 23, 2001, all of which are herein incorporated in their entirety. This application also claims priority to U.S. provisional 60/476,194 filed on Jun. 6, 2003; U.S. provisional 60/504,731 filed on Sep. 22, 2003; U.S. provisional 60/528,249 filed on Dec. 10, 2003; U.S. provisional 60/557,671 filed on Mar. 21, 2004 and U.S. provisional 60/(awaited) filed on Jun. 2, 2004, all of which are herein incorporated in their entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to apoptosis-specific eucaryotic initiation factor (“eIF-5A”) or referred to as “apoptosis-specific eIF-5A” or “eIF-5A1” and deoxyhypusine synthase (DHS). The present invention relates to apoptosis-specific eIF-5A and DHS nucleic acids and polypeptides and methods for inhibiting expression of apoptosis-specific eIF-5A and DHS.
  • BACKGROUND OF THE INVENTION
  • Apoptosis is a genetically programmed cellular event that is characterized by well-defined morphological features, such as cell shrinkage, chromatin condensation, nuclear fragmentation, and membrane blebbing. Kerr et al. (1972) Br. J. Cancer, 26, 239-257; Wyllie et al. (1980) Int. Rev. Cytol., 68, 251-306. It plays an important role in normal tissue development and homeostasis, and defects in the apoptotic program are thought to contribute to a wide range of human disorders ranging from neurodegenerative and autoimmunity disorders to neoplasms. Thompson (1995) Science, 267, 1456-1462; Mullauer et al. (2001) Mutat. Res, 488, 211-231. Although the morphological characteristics of apoptotic cells are well characterized, the molecular pathways that regulate this process have only begun to be elucidated.
  • One group of proteins that is thought to play a key role in apoptosis is a family of cysteine proteases, termed caspases, which appear to be required for most pathways of apoptosis. Creagh & Martin (2001) Biochem. Soc. Trans, 29, 696-701; Dales et al. (2001) Leuk. Lymphoma, 41, 247-253. Caspases trigger apoptosis in response to apoptotic stimuli by cleaving various cellular proteins, which results in classic manifestations of apoptosis, including cell shrinkage, membrane blebbing and DNA fragmentation. Chang & Yang (2000) Microbiol. Mol. Biol. Rev., 64, 821-846.
  • Pro-apoptotic proteins, such as Bax or Bak, also play a key role in the apoptotic pathway by releasing caspase-activating molecules, such as mitochondrial cytochrome c, thereby promoting cell death through apoptosis. Martinou & Green (2001) Nat. Rev. Mol. Cell. Biol., 2, 63-67; Zou et al. (1997) Cell, 90, 405-413. Anti-apoptotic proteins, such as Bcl-2, promote cell survival by antagonizing the activity of the pro-apoptotic proteins, Bax and Bak. Tsujimoto (1998) Genes Cells, 3, 697-707; Kroemer (1997) Nature Med., 3, 614-620. The ratio of Bax:Bcl-2 is thought to be one way in which cell fate is determined; an excess of Bax promotes apoptosis and an excess of Bcl-2 promotes cell survival. Salomons et al. (1997) Int. J. Cancer, 71, 959-965; Wallace-Brodeur & Lowe (1999) Cell Mol. Life Sci., 55, 64-75.
  • Another key protein involved in apoptosis is a protein that encoded by the tumor suppressor gene p53. This protein is a transcription factor that regulates cell growth and induces apoptosis in cells that are damaged and genetically unstable, presumably through up-regulation of Bax. Bold et al. (1997) Surgical Oncology, 6, 133-142; Ronen et al., 1996; Schuler & Green (2001) Biochem. Soc. Trans., 29, 684-688; Ryan et al. (2001) Curr. Opin. Cell Biol., 13, 332-337; Zörmig et al. (2001) Biochem. Biophys. Acta, 1551, F1-F37.
  • Alterations in the apoptotic pathways are believed to play a key role in a number of disease processes, including cancer. Wyllie et al. (1980) Int. Rev. Cytol., 68, 251-306; Thompson (1995) Science, 267, 1456-1462; Sen & D'Incalci (1992) FEBS Letters, 307, 122-127; McDonnell et al. (1995) Seminars in Cancer and Biology, 6, 53-60. Investigations into cancer development and progression have traditionally been focused on cellular proliferation. However, the important role that apoptosis plays in tumorigenesis has recently become apparent. In fact, much of what is now known about apoptosis has been learned using tumor models, since the control of apoptosis is invariably altered in some way in tumor cells. Bold et al. (1997) Surgical Oncology, 6, 133-142.
  • Cytokines also have been implicated in the apoptotic pathway. Biological systems require cellular interactions for their regulation, and cross-talk between cells generally involves a large variety of cytokines. Cytokines are mediators that are produced in response to a wide variety of stimuli by many different cell types. Cytokines are pleiotropic molecules that can exert many different effects on many different cell types, but are especially important in regulation of the immune response and hematopoietic cell proliferation and differentiation. The actions of cytokines on target cells can promote cell survival, proliferation, activation, differentiation, or apoptosis depending on the particular cytokine, relative concentration, and presence of other mediators.
  • The use of anti-cytokines to treat autoimmune disorders such as psoriasis, rheumatoid arthritis, and Crohn's disease is gaining popularity. The pro-inflammatory cytokines IL-1 and TNF play a large role in the pathology of these chronic disorders. Anti-cytokine therapies that reduce the biological activities of these two cytokines can provide therapeutic benefits (Dinarello and Abraham, 2002).
  • Interleukin 1 (IL-I) is an important cytokine that mediates local and systemic inflammatory reactions and which can synergize with TNF in the pathogenesis of many disorders, including vasculitis, osteoporosis, neurodegenerative disorders, diabetes, lupus nephritis, and autoimmune disorders such as rheumatoid arthritis. The importance of IL-1β in tumour angiogenesis and invasiveness was also recently demonstrated by the resistance of IL-1β knockout mice to metastases and angiogenesis when injected with melanoma cells (Voronov et al., 2003).
  • Interleukin 18 (IL-18) is a recently discovered member of the IL-1 family and is related by structure, receptors, and function to IL-1. IL-18 is a central cytokine involved in inflammatory and autoimmune disorders as a result of its ability to induce interferon-gamma (IFN-γ), TNF-α, and IL-1. IL-1β and IL-18 are both capable of inducing production of TNF-α, a cytokine known to contribute to cardiac dysfunction during myocardial ischemia (Maekawa et al., 2002). Inhibition of IL-18 by neutralization with an IL-18 binding protein was found to reduce ischemia-induced myocardial dysfunction in an ischemia/reperfusion model of suprafused human atrial myocardium (Dinarello, 2001). Neutralization of IL-18 using a mouse IL-18 binding protein was also able to decrease IFN-γ, TNF-α, and IL-1β transcript levels and reduce joint damage in a collagen-induced arthritis mouse model (Banda et al., 2003). A reduction of IL-18 production or availability may also prove beneficial to control metastatic cancer as injection of IL-18 binding protein in a mouse melanoma model successfully inhibited metastases (Carrascal et al., 2003). As a further indication of its importance as a pro-inflammatory cytokine, plasma levels of IL-18 were elevated in patients with chronic liver disease and increased levels were correlated with the severity of the disease (Ludwiczek et aL., 2002). Similarly, IL-18 and TNF-α were elevated in the serum of diabetes mellitus patients with nephropathy (Moriwaki et al., 2003). Neuroinflammation following traumatic brain injury is also mediated by pro-inflammatory cytokines and inhibition of IL-18 by the IL-18 binding protein improved neurological recovery in mice following brain trauma (Yatsiv et al., 2002).
  • TNF-α, a member of the TNF family of cytokines, is a pro-inflammatory cytokine with pleiotropic effects ranging from co-mitogenic effects on hematopoietic cells, induction of inflammatory responses, and induction of cell death in many cell types. TNF-α is normally induced by bacterial lipopolysaccharides, parasites, viruses, malignant cells and cytokines and usually acts beneficially to protect cells from infection and cancer. However, inappropriate induction of TNF-α is a major contributor to disorders resulting from acute and chronic inflammation such as autoimmune disorders and can also contribute to cancer, AIDS, heart disease, and sepsis (reviewed by Aggarwal and Natarajan, 1996; Sharma and Anker, 2002). Experimental animal models of disease (i.e. septic shock and rheumatoid arthritis) as well as human disorders (i.e. inflammatory bowel diseases and acute graft-versus-host disease) have demonstrated the beneficial effects of blocking TNF-α (Wallach et al., 1999). Inhibition of TNF-α has also been effective in providing relief to patients suffering autoimmune disorders such as Crohn's disease (van Deventer, 1999) and rheumatoid arthritis (Richard-Miceli and Dougados, 2001). The ability of TNF-α to promote the survival and growth of B lymphocytes is also thought to play a role in the pathogenesis of B-cell chronic lymphocytic leukemia (B-CLL) and the levels of TNF-α being expressed by T cells in B-CLL was positively correlated with tumour mass and stage of the disease (Bojarska-Junak et al., 2002). Interleukin-1β (IL-1β) is a cytokine known to induce TNF-α production.
  • Thus, since the accumulation of excess cytokines and TNF-α can lead to deleterious consequences on the body, including cell death, there is a need for a method to reduce the levels of cytokines in the body as well as inhibiting or reducing apoptosis. The present invention fulfills these needs.
  • Deoxyhypusine synthase (DHS) and hypusine-containing eucaryotic translation initiation Factor-5A (eIF-5A) are known to play important roles in many cellular processes including cell growth and differentiation. Hypusine, a unique amino acid, is found in all examined eucaryotes and archaebacteria, but not in eubacteria, and eIF-5A is the only known hypusine-containing protein. Park (1988) J. Biol. Chem., 263, 7447-7449; Schüimann & Klink (1989) System. Appl. Microbiol., 11, 103-107; Bartig et al. (1990) System. Appl. Microbiol., 13, 112-116; Gordon et al. (1987a) J. Biol. Chem., 262, 16585-16589. Active eIF-5A is formed in two post-translational steps: the first step is the formation of a deoxyhypusine residue by the transfer of the 4-aminobutyl moiety of spermidine to the α-amino group of a specific lysine of the precursor eIF-5A catalyzed by deoxyhypusine synthase; the second step involves the hydroxylation of this 4-aminobutyl moiety by deoxyhypusine hydroxylase to form hypusine.
  • The amino acid sequence of eIF-5A is well conserved between species, and there is strict conservation of the amino acid sequence surrounding the hypusine residue in eIF-5A, which suggests that this modification may be important for survival. Park et al. (1993) Biofactors, 4, 95-104. This assumption is further supported by the observation that inactivation of both isoforms of eIF-5A found to date in yeast, or inactivation of the DHS gene, which catalyzes the first step in their activation, blocks cell division. Schnier et al. (1991) Mol. Cell. Biol., 11, 3105-3114; Sasaki et al. (1996) FEBS Lett., 384, 151-154; Park et al. (1998) J. Biol. Chem., 273, 1677-1683. However, depletion of eIF-5A protein in yeast resulted in only a small decrease in total protein synthesis suggesting that eIF-5A may be required for the translation of specific subsets of mRNA's rather than for protein global synthesis. Kang et al. (1993), “Effect of initiation factor eIF-5A depletion on cell proliferation and protein synthesis,” in Tuite, M. (ed.), Protein Synthesis and Targeting in Yeast, NATO Series H. The recent finding that ligands that bind eIF-5A share highly conserved motifs also supports the importance of eIF-5A. Xu & Chen (2001) J. Biol. Chem., 276, 2555-2561. In addition, the hypusine residue of modified eIF-5A was found to be essential for sequence-specific binding to RNA, and binding did not provide protection from ribonucleases.
  • In addition, intracellular depletion of eIF-5A results in a significant accumulation of specific mRNAs in the nucleus, indicating that eIF-5A may be responsible for shuttling specific classes of mRNAs from the nucleus to the cytoplasm. Liu & Tartakoff (1997) Supplement to Molecular Biology of the Cell, 8, 426a. Abstract No. 2476, 37th American Society for Cell Biology Annual Meeting. The accumulation of eIF-5A at nuclear pore-associated intranuclear filaments and its interaction with a general nuclear export receptor further suggest that eIF-5A is a nucleocytoplasmic shuttle protein, rather than a component of polysomes. Rosorius et al. (1999) J. Cell Science, 112, 2369-2380.
  • The first cDNA for eIF-5A was cloned from human in 1989 by Smit-McBride et al., and since then cDNAs or genes for eIF-5A have been cloned from various eukaryotes including yeast, rat, chick embryo, alfalfa, and tomato. Smit-McBride et al. (1989) J. Biol. Chem., 264, 1578-1583; Schnier et al. (1991) (yeast); Sano, A. (1995) in Imahori, M. et al. (eds), Polyamines, Basic and Clinical Aspects, VNU Science Press, The Netherlands, 81-88 (rat); Rinaudo & Park (1992) FASEB J., 6, A453 (chick embryo); Pay et al. (1991) Plant Mol. Biol., 17, 927-929 (alfalfa); Wang et al. (2001) J. Biol. Chem., 276, 17541-17549 (tomato).
  • Expression of eIF-5A mRNA has been explored in various human tissues and mammalian cell lines. For example, changes in eIF-5A expression have been observed in human fibroblast cells after addition of serum following serum deprivation. Pang & Chen (1994) J. Cell Physiol., 160, 531-538. Age-related decreases in deoxyhypusine synthase activity and abundance of precursor eIF-5A have also been observed in senescing fibroblast cells, although the possibility that this reflects averaging of differential changes in isoforms was not determined. Chen & Chen (1997) J. Cell Physiol., 170, 248-254.
  • Studies have shown that eIF-5A may be the cellular target of viral proteins such as the human immunodeficiency virus type 1 Rev protein and human T cell leukemia virus type 1 Rex protein. Ruhl et al. (1993) J. Cell Biol.,123, 1309-1320; Katahira et al. (1995) J. Virol., 69, 3125-3133. Preliminary studies indicate that eIF-5A may target RNA by interacting with other RNA-binding proteins such as Rev, suggesting that these viral proteins may recruit eIF-5A for viral RNA processing. Liu et al. (1997) Biol. Signals, 6, 166-174.
  • Thus, although eIF5A and DHS are known, there remains a need in understanding how these proteins are involved in apoptotic pathways as well as cytokine stimulation to be able to modulate apoptosis and cytokine expression. The present invention fulfills this need.
  • SUMMARY OF INVENTION
  • The present invention relates to apoptosis specific eucaryotic initiation factor 5A (eIF-5A), referred to as “apoptosis specific eIF-5A” or “eIF-5A1.” The present invention also relates to apoptosis-specific eIF-5A nucleic acids and polypeptides and methods for inhibiting or suppressing apoptosis in cells using antisense nucleotides or siRNAs to inhibit expression of apoptosis-specific eIF-5A. The present invention relates to a method of delivering siRNA to mammalian lung cells in vivo. The invention also relates to suppressing or inhibiting expression of pro-inflammatory cytokines by inhibiting expression of apoptosis-specific eIF-5A. Further, the present invention relates to inhibiting or suppressing expression of p53 by inhibiting expression of apoptosis-specific eIF-5A. The present invention also relates to a method of increasing Bcl-2 expression by inhibiting or suppression expression of apoptosis factor 5A using antisense nucleotides or siRNAs. The present invention also provides a method of inhibiting production of cytokines, especially TNF-α in human epithelial cells. In another embodiment of the present invention, suppressing expression of apoptosis-specific eIF-5A by the use of antisense oligonucleotides targeted at apoptosis-specific eIF-5A provides methods of preventing retinal ganglion cell death in a glaucomatous eye.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts the nucleotide sequence (SEQ ID NO: 11) and derived amino acid sequence (SEQ ID NO: 12) of the 3′ end of rat apoptosis-specific eIF-5A.
  • FIG. 2 depicts the nucleotide sequence (SEQ ID NO: 15) and derived amino acid sequence (SEQ ID NO: 16) of the 5′ end of rat apoptosis-specific eIF-5A cDNA.
  • FIG. 3 depicts the nucleotide sequence of rat corpus luteum apoptosis-specific eIF-5A full-length cDNA (SEQ ID NO: 1). The amino acid sequence is shown in SEQ ID NO: 2.
  • FIG. 4 depicts the nucleotide sequence (SEQ ID NO: 6) and derived amino acid sequence (SEQ ID NO: 7) of the 3′ end of rat apoptosis-specific DHS cDNA.
  • FIG. 5 is an alignment of the full-length nucleotide sequence of rat corpus luteum apoptosis-specific eIF-5A cDNA (SEQ ID NO: 20) with the nucleotide sequence of human eIF-5A (SEQ ID NO: 3) (Accession number BC000751 or NM001970, SEQ ID NO:3).
  • FIG. 6 is an alignment of the full-length nucleotide sequence of rat corpus luteum apoptosis-specific eIF-5A cDNA (SEQ ID NO: 20) with the nucleotide sequence of human eIF-5A (SEQ ID NO: 4) (Accession number NM-020390, SEQ ID NO:4).
  • FIG. 7 is an alignment of the full-length nucleotide sequence of rat corpus luteum apoptosis-specific eIF-5A cDNA (SEQ ID NO: 20) with the nucleotide sequence of mouse eIF-5A (Accession number BC003889). Mouse nucleotide sequence (Accession number BC003889) is SEQ ID NO:5.
  • FIG. 8 is an alignment of the derived full-length amino acid sequence of rat corpus luteum apoptosis-specific eIF-5A (SEQ ID NO: 2) with the derived amino acid sequence of human eIF-5A (SEQ ID NO: 21) (Accession number BC000751 or NM001970).
  • FIG. 9 is an alignment of the derived full-length amino acid sequence of rat corpus luteum apoptosis-specific eIF-5A (SEQ ID NO: 2) with the derived amino acid sequence of human eIF-5A (SEQ ID NO: 22) (Accession number NM020390).
  • FIG. 10 is an alignment of the derived full-length amino acid sequence of rat corpus luteum apoptosis-specific eIF-5A (SEQ ID NO: 2) with the derived amino acid sequence of mouse eIF-5A (SEQ ID NO: 23) (Accession number BC003889).
  • FIG. 11 is an alignment of the partial-length nucleotide sequence of rat corpus luteum apoptosis-specific DHS cDNA (residues 1-453 of SEQ ID NO: 6) with the nucleotide sequence of human DHS (SEQ ID NO: 8) (Accession number BC000333, SEQ ID NO:8).
  • FIG. 12 is a restriction map of rat corpus luteum apoptosis-specific eIF-5A cDNA.
  • FIG. 13 is a restriction map of the partial-length rat apoptosis-specific DHS cDNA.
  • FIG. 14 is a Northern blot (top) and an ethidium bromide stained gel (bottom) of total RNA probed with the 32P-dCTP-labeled 3′-end of rat corpus luteum apoptosis-specific eIF-5A cDNA.
  • FIG. 15 is a Northern blot (top) and an ethidium bromide stained gel (bottom) of total RNA probed with the 32P-dCTP-labeled 3′-end of rat corpus luteum apoptosis-specific DHS cDNA.
  • FIG. 16 depicts a DNA laddering experiment in which the degree of apoptosis in superovulated rat corpus lutea was examined after injection with PGF-2α.
  • FIG. 17 is an agarose gel of genomic DNA isolated from apoptosing rat corpus luteum showing DNA laddering after treatment of rats with PGF F-2α.
  • FIG. 18 depicts a DNA laddering experiment in which the degree of apoptosis in dispersed cells of superovulated rat corpora lutea was examined in rats treated with sperrnidine prior to exposure to PGF-2α.
  • FIG. 19 depicts a DNA laddering experiment in which the degree of apoptosis in superovulated rat corpus lutea was examined in rats treated with spermidine and/or PGF-2α.
  • FIG. 20 is a Southern blot of rat genomic DNA probed with 32P-dCTP-labeled partial-length rat corpus luteum apoptosis-specific eIF-5A cDNA.
  • FIG. 21 depicts pHM6, a mammalian epitope tag expression vector (Roche Molecular Biochemicals).
  • FIG. 22 is a Northern blot (top) and ethidium bromide stained gel (bottom) of total RNA isolated from COS-7 cells after induction of apoptosis by withdrawal of serum probed with the 32P-dCTP-labeled 3′-untranslated region of rat corpus luteum apoptosis-specific DHS cDNA.
  • FIG. 23 is a flow chart illustrating the procedure for transient transfection of COS-7 cells.
  • FIG. 24 is a Western blot of transient expression of foreign proteins in COS-7 cells following transfection with pHM6.
  • FIG. 25 illustrates enhanced apoptosis as reflected by increased caspase activity when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 26 illustrates enhanced apoptosis as reflected by increased DNA fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 27 illustrates detection of apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 28 illustrates enhanced apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 29 illustrates detection of apoptosis as reflected by phosphatidylserine exposure when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 30 illustrates enhanced apoptosis as reflected by increased phosphatidylserine exposure when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 31 illustrates enhanced apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 32 illustrates enhanced apoptosis when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation.
  • FIG. 33 illustrates down-regulation of Bcl-2 when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation. The top photo is the Coomassie-blue-stained protein blot; the bottom photo is the corresponding Western blot.
  • FIG. 34 is a Coomassie-blue-stained protein blot and the corresponding Western blot of COS-7 cells transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the antisense orientation using Bcl-2 as a probe.
  • FIG. 35 is a Coomassie-blue-stained protein blot and the corresponding Western blot of COS-7 cells transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation using c-Myc as a probe.
  • FIG. 36 is a Coomassie-blue-stained protein blot and the corresponding Western blot of COS-7 cells transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation when p53 is used as a probe.
  • FIG. 37 is a Coomassie-blue-stained protein blot and the corresponding Western blot of expression of pHM6-full-length rat apoptosis-specific eIF-5A in COS-7 cells using an anti-[HA]-peroxidase probe and a Coomassie-blue-stained protein blot and the corresponding Western blot of expression of pHM6-full-length rat apoptosis-specific eIF-5A in COS-7 cells when a p53 probe is used.
  • FIG. 38 is an alignment of human eIF5A2 isolated from RKO cells (SEQ ID NO: 24) with the sequence of human eIF5A2 (SEQ ID NO: 22) (Genbank accession number XM113401). The consensus sequence is shown in SEQ ID NO: 28.
  • FIG. 39 is a graph depicting the percentage of apoptosis occurring in RKO and RKO-E6 cells following transient transfection. RKO and RKO-E6 cells were transiently transfected with pHM6-LacZ or pHM6-apoptosis-specific eIF-5A. RKO cells treated with Actinomycin D and transfected with pHM6-apoptosis-specific eIF-5A showed a 240% increase in apoptosis relative to cells transfected with pHM6-LacZ that were not treated with Actinomycin D. RKO-E6 cells treated with Actinomycin D and transfected with pHM6-apoptosis-specific eIF-5A showed a 105% increase in apoptosis relative to cells transfected with pHM6-LacZ that were not treated with Actinomycin D.
  • FIG. 40 is a graph depicting the percentage of apoptosis occurring in RKO cells following transient transfection. RKO cells were transiently transfected with pHM6-LacZ, pHM6-apoptosis-specific eIF-5A, pHM6-eIF5A2, or pHM6-truncated apoptosis-specific eIF-5A. Cells transfected with pHM6-apoptosis-specific eIF-5A showed a 25% increase in apoptosis relative to control cells transfected with pHM6-LacZ. This increase was not apparent for cells transfected with pHM6-eIF5A2 or pHM6-truncated apoptosis-specific eIF-5A.
  • FIG. 41 is a graph depicting the percentage of apoptosis occurring in RKO cells following transient transfection. RKO cells were either left untransfected or were transiently transfected with pHM6-LacZ or pHM6-apoptosis-specific eIF-5A. After correction for transfection efficiency, 60% of the cells transfected with pHM6-apoptosis-specific eIF-5A were apoptotic.
  • FIG. 42 provides the results of a flow cytometry analysis of RKO cell apoptosis following transient transfection. RKO cells were either left untransfected or were transiently transfected with pHM6-LacZ, pHM6-apoptosis-specific eIF-5A, pHM6-eIF5A2, or pHM6-truncated apoptosis-specific eIF-5A. The table depicts the percentage of cells undergoing apoptosis calculated based on the area under the peak of each gate. After correction for background apoptosis in untransfected cells and for transfection efficiency, 80% of cells transfected with pHM6-apoptosis-specific eIF-5A exhibited apoptosis. Cells transfected with pHM6-LacZ, pHM6-eIF5A2 or pHM6-truncated apoptosis-specific eIF-5A exhibited only background levels of apoptosis.
  • FIG. 43 provides Western blots of protein extracted from RKO cells treated with 0.25 μg/ml Actinomycin D for 0, 3, 7, 24, and 48 hours. The top panel depicts a Western blot using anti-p53 as the primary antibody. The middle panel depicts a Western blot using anti-apoptosis-specific eIF-5A as the primary antibody. The bottom panel depicts the membrane used for the anti-apoptosis-specific eIF-5A blot stained with Coomassie blue following chemiluminescent detection to demonstrate equal loading. p53 and apoptosis-specific eIF-5A are both upregulated by treatment with Actinomycin D.
  • FIG. 44 is a bar graph showing that both apoptosis-specific eIF-5A and proliferation eIF-5A are expressed in heart tissue. The heart tissue was taken from patients receiving coronary artery bypass grafts (“CABG”). Gene expression levels apoptosis-specific eIF-5A (light gray bar) are compared to proliferation eIF-5A (dark gray bar). The X-axis is patient identifier numbers. The Y-axis is pg/ng of 18s (picograms of message RNA over nanograms of ribosomal RNA 18S).
  • FIG. 45 is a bar graph showing that both apoptosis-specific eIF-5A and proliferation eIF-5A are expressed in heart tissue. The heart tissue was taken from patients receiving valve replacements. Gene expression levels of apoptosis-specific eIF-5A (light gray bar) are compared to proliferation eIF-5A (dark gray bar). The X-axis is patient identifier numbers. The Y-axis is pg/ng of 18s (picograms of message RNA over nanograms of ribosomal RNA 18S).
  • FIG. 46 is a bar graph showing the gene expression levels measured by real-time PCR of apoptosis-specific eIF-5A (elf5a) versus proliferation eIF-5A (eIF5b) in pre-ischemia heart tissue and post ischemia heart tissue. The Y-axis is pg/ng of 18s (picograms of message RNA over nanograms of ribosomal RNA 18S).
  • FIG. 47 depicts schematically an experiment performed on heart tissue. The heart tissue was exposed to normal oxygen levels and the expression levels apoptosis-specific eIF-5A and proliferating eIF-5A measured. Later, the amount of oxygen delivered to the heart tissue was lowered, thus inducing hypoxia and ischemia, and ultimately, a heart attack in the heart tissue. The expression levels of apoptosis-specific eIF-5A and proliferating eIF-5A were measured and compared to the expression levels of the heart tissue before it was damaged by ischemia.
  • FIG. 48 shows EKGs of heart tissue before and after the ischemia was induced.
  • FIG. 49 shows the lab bench with the set up of the experiment depicted in FIG. 47.
  • FIGS. 50A-F report patient data where the levels of apoptosis-specific eIF-5A are correlated with levels of IL-1β and IL-18. FIG. 50A is a chart of data obtained from coronary artery bypass graft (CABG) patients. FIG. 50B is a chart of data obtained from valve replacement patients. FIG. 50C is a graph depicting the correlation of apoptosis-specific eIF-5A to IL-18 in CABG patients. FIG. 50D is a graph depicting the correlation of proliferating eIF-5A to IL-18 in CABG patients. FIG. 50E is a graph depicting the correlation of apoptosis-specific eIF-5A to IL-18 in valve replacement patients. FIG. 50F is a graph depicting the correlation of proliferating eIF-5A to IL-18 in valve replacement patients.
  • FIG. 51 is a chart of the patient's data from which patients data used in FIGS. 50A-F was obtained.
  • FIG. 52 shows the levels of protein produced by RKO cells after being treated with antisense oligo 1, 2 and 3 (of apoptosis-specific eIF-5A)(SEQ ID NO: 35, 37 and 39, respectively). The RKO cells produced less apoptosis-specific eIF-5A as well as less p53 after having been transfected with the antisense apoptosis-specific eIF-5A oligonucleotides.
  • FIG. 53 shows uptake of the fluorescently labeled antisense oligonucleotide.
  • FIGS. 54-58 show a decrease in the percentage of cells undergoing apoptosis in the cells having being treated with antisense apoptosis-specific eIF-5A oligonucleotides as compared to cells not having been transfected with the antisense apoptosis-specific eIF-5A oligonucleotides.
  • FIG. 59 shows that treating lamina cribrosa cells with TNF-α and/or camptothecin caused an increase in the number of cells undergoing apoptosis.
  • FIGS. 60 and 61 shows a decrease in the percentage of cells undergoing apoptosis in the cells having being treated with antisense apoptosis-specific eIF-5A oligonucleotides as compared to cells not having been transfected with the antisense apoptosis-specific eIF-5A oligonucleotides.
  • FIG. 62 shows that the lamina cribrosa cells uptake the labeled siRNA either in the presence of serum or without serum.
  • FIG. 63 shows that cells transfected with apoptosis-specific eIF-5A siRNA produced less apoptosis-specific eIF-5A protein and in addition, produced more Bcl-2 protein. A decrease in apoptosis-specific eIF-5A expression correlates with an increase in BCL-2 expression.
  • FIG. 64 shows that cells transfected with apoptosis-specific eIF-5A siRNA produced less apoptosis factor 5a protein.
  • FIGS. 65-67 shows that cells transfected with apoptosis-specific eIF-5A siRNA had a lower percentage of cells undergoing apoptosis after exposure to amptothecin and TNF-α than untransfected cells.
  • FIG. 68 are photographs of Hoescht-stained lamina cribrosa cell line #506 transfected with siRNA and treated with camptothecin and TNF-α from the experiment described in
  • FIG. 67 and Example 13. The apoptosing cells are seen as more brightly stained cells. They have smaller nucleic because of chromatin condensation and are smaller and irregular in shape.
  • FIG. 69 shows that IL-1 exposed HepG2 cells transfected with apoptosis-specific eIF-5A cells secreted less TNF-α than non-transfected cells.
  • FIG. 70 shows the sequence of human apoptosis-specific eIF-5A (SEQ ID NO:29) and the sequences of 5 siRNAs of the present invention (SEQ ID NO:30, 31, 32, 33 and 34).
  • FIG. 71 shows the sequence of human apoptosis-specific eIF-5A (SEQ ID NO: 29) and the sequences of 3 antisense oligonucleotides of the present invention (SEQ ID NO:35, 37, and 39, respectively in order of appearance ).
  • FIG. 72 shows the binding position of three antisense oligonucleotides (SEQ ID NO:25-27, respectively in order of appearance) targeted against human apoptosis-specific eIF-5A. The full-length nucleotide sequence is SEQ ID NO: 19.
  • FIG. 73 a and b shows the nucleotide alignment (SEQ ID NO: 41 and 42, respectively in order of appearance) and amino acid alignment (SEQ ID NO: 43 and 22, respectively in order of appearance) of human apoptosis-specific eIF-5A against human proliferating eIF-5A.
  • FIG. 74A provides a picture of a Western blot where siRNAs against apoptosis-specific eIF-5A have reduced if not inhibited the production of TNF-α in transfected HT-29 cells.
  • FIG. 74B provides the results of an ELISA.
  • FIG. 75 provides the results of an ELISA. TNF-α production was reduced in cells treated with siRNAs against apoptosis-specific eIF-5A as compared to control cells.
  • FIG. 76 shows the time course of the U-937 differentiation experiment. See Example 16.
  • FIG. 77 shows the results of a Western blot showing that apoptosis-specific eIF-5A is up-regulated during monocyte differentiation and subsequence TNF-α secretion.
  • FIG. 78 depicts stem cell differentiation and the use of siRNAs against apoptosis-specific eIF-5A to inhibit cytokine production.
  • FIG. 79 is a bar graph showing that IL-8 is produced in response to TNF-alpha as well as in response to interferon. This graph shows that siRNA against apoptosis-specific eIF-5A blocked almost all IL-8 produced in response to interferon as well as a significant amount of the IL-8 produced as a result of the combined treatment of interferon and TNF.
  • FIG. 80 is another bar graph showing that IL-8 is produced in response to TNF-alpha as well as in response to interferon. This graph shows that siRNA against apoptosis-specific eIF-5A blocked almost all IL-8 produced in response to interferon as well as a significant amount of the IL-8 produced as a result of the combined treatment of interferon and TNF.
  • FIG. 81 is a western blot of HT-29 cells treated with IFN gamma for 8 and 24 hours. This blot shows up-regulation in HT-29 cells (4 fold at 8 hours) of against apoptosis-specific eIF-5A in response to interferon gamma.
  • FIG. 82 is a characterization of lamina cribrosa cells by immunofluorescence. Lamina cribrosa cells (#506) isolated from the optic nerve head of an 83-year old male were characterized by immunofluorescence. Primary antibodies were a) actin; b) fibronectin; c) laminin; d) GFAP. All pictures were taken at 400 times magnification.
  • FIG. 83 is a graph showing percent apoptosis of lamina cribrosa cell line #506 in response to treatment with camptothecin and TNF-α. Lamina cribrosa cell line #506 cells were seeded at 40,000 cells per well onto an 8-well culture slide. Three days later the confluent LC cells were treated with either 10 ng/ml TNF-α, 50 μM camptothecin, or 10 ng/ml TNF-α plus 50 μM camptothecin. An equivalent volume of DMSO, a vehicle control for camptothecin, was added to the untreated control cells. The cells were stained with Hoescht 33258 48 hours after treatment and viewed by fluorescence microscopy using a UV filter. Cells with brightly stained condensed or fragmented nuclei were counted as apoptotic.
  • FIG. 84 shows expression levels of against apoptosis-specific eIF-5A during camptothecin or TNF-a plus camptothecin treatment. Lamina cribrosa cell #506 cells were seeded at 40,000 cells per well onto a 24-well plate. Three days later the LC cells were treated with either 50 μM camptothecin or 10 ng/ml TNF-α plus 50 μM camptothecin and protein lysate was harvested 1, 4, 8, and 24 hours later. An equivalent volume of DMSO was added to control cells as a vehicle control and cell lysate was harvested 1 and 24 hours later. 5 μg of protein from each sample was separated by SDS-PAGE, transferred to a PVDF membrane, and Western blot with anti-apoptosis-specific eIF-5A antibody. The bound antibody was detected by chemiluminescence and exposed to x-ray film. The membrane was then stripped and re-blotted with anti-β-actin as an internal loading control.
  • FIG. 85 shows expression levels of apoptosis-specific eIF-5A in lamina cribosa cell lines #506 and #517 following transfection with siRNAs. Lamina cribrosa cell #506 and #517 cells were seeded at 10,000 cells per well onto a 24-well plate. Three days later the LC cells were transfected with either GAPDH siRNA, apoptosis-specific eIF-5A siRNAs #1-4 (SEQ ID NO:30-33) or control siRNA #5 (SEQ ID NO:34). Three days after transfection the protein lysate was harvested and 5 μg of protein from each sample was separated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with anti-eIF5A antibody. The bound antibody was detected by chemiluminescence and exposed to x-ray film. The membrane was then stripped and re-blotted with anti-β-actin as an internal loading control. This figure shows that cells treated with siRNAs of apoptosis-specific eIF-5A produce less apoptosis-specific eIF-5A protein.
  • FIG. 86 shows the percent apoptosis of lamina cribosa cell line #506 cells transfected with apoptosis-specific eIF-5A siRNAs and treated with TNF-α and camptothecin. Lamina cribrosa cell line #506 cells were seeded at 7500 cells per well onto an 8-well culture slide. Three days later the LC cells were transfected with either GAPDH siRNA, apoptosis-specific eIF-5A siRNAs #1-4 (SEQ ID NO:30-33), or control siRNA #5 (SEQ ID NO:34). 72 hours after transfection, the transfected cells were treated with 10 ng/ml TNF-α plus 50 μM camptothecin. Twenty-four hours later the cells were stained with Hoescht 33258 and viewed by fluorescence microscopy using a UV filter. Cells with brightly stained condensed or fragmented nuclei were counted as apoptotic. This graph represents the average of n=4 independent experiments. This figure shows that cells treated with siRNAs of apoptosis-specific eIF-5A show a smaller percentage of apoptosis upon treatment with camptothecin and TNF as compared to cells not transfected with siRNAs of apoptosis-specific eIF-5A.
  • FIG. 87 shows percent apoptosis of lamina cribosa cell line #517 cells transfected with apoptosis-specific eIF-5A siRNA #1 and treated with TNF-α and camptothecin. Lamina cribrosa cell line #517 cells were seeded at 7500 cells per well onto an 8-well culture slide. Three days later the LC cells were transfected with either apoptosis-specific eIF-5A siRNA #1 (SEQ ID NO:30) or control siRNA #5 (SEQ ID NO:34). 72 hours after transfection, the transfected cells were treated with 10 ng/ml TNF-α plus 50 μM camptothecin. Twenty-four hours later the cells were stained with Hoescht 33258 and viewed by fluorescence microscopy using a UV filter. Cells with brightly stained condensed or fragmented nuclei were counted as apoptotic. The results of two independent experiments are represented here. This shows that cells treated with siRNAs had a lower percentage of apoptosis.
  • FIG. 88 shows TUNEL-labeling of lamina cribosa cell line #506 cells transfected with apoptosis-specific eIF-5A siRNA #1 and treated with TNF-α and camptothecin. Lamina cribrosa cell line #506 cells were seeded at 7500 cells per well onto an 8-well culture slide. Three days later the LC cells were transfected with either apoptosis-specific eIF-5A siRNA #1 (SEQ ID NO:30) or control siRNA #5 (SEQ ID NO:34). 72 hours after transfection, the transfected cells were treated with 10 ng/ml TNF-α plus 50 μM camptothecin. Twenty-four hours later the cells were stained with Hoescht 33258 and DNA fragmentation was evaluated in situ using the terminal deoxynucleotidyl transferase-mediated dUTP-digoxigenin nick end labeling (TUNEL) method. Panel A represents the slide observed by fluorescence microscopy using a fluorescein filter to visualize TUNEL-labeling of the fragmented DNA of apoptotic cells. Panel B represents the same slide observed by through a UV filter to visualize the Hoescht-stained nuclei. The results are representative of two independent experiments. All pictures were taken at 400 times magnification.
  • FIG. 89 depicts the design of siRNAs against apoptosis-specific eIF-5A. The siRNAs have the SEQ ID NO: 45, 48, 51, 54 and 56. The full-length nucleotide sequence is shown in SEQ ID NO: 29.
  • FIG. 90 shows the results of an experiment where siRNAs directed against apoptosis-specific eIF-5A provided for a reduction in NKkB activation in the presence of interferon gamma and LPS.
  • FIG. 91 shows the time course for PBMC experiments (see Example 18).
  • FIG. 92 shows a Western blot of a cell lysate from PBMCs collected from two donors over a time course. The PBMCs were treated with PMA and subsequently stimulated with LPS to have an increased apoptosis-specific eIF-5A expression.
  • FIG. 93 shows that PBMCs treated with PMA and subsequently stimulated with LPS have an increased apoptosis-specific eIF-5A expression, which coincides with increased TNF production.
  • FIG. 94 demonstrates that PBMCs respond to LPS without PMA differentiation.
  • FIG. 95 shows that PBMCs transfected with apoptosis-specific eIF-5A siRNAs demonstrate suppression of expression of apoptosis-specific eIF-5A.
  • FIG. 96 shows that PBMCs transfected with apoptosis-specific eIF-5A siRNAs and stimulated with LPS produce less TNF than PBMCs not transfected with apoptosis-specific eIF-5A siRNAs.
  • FIG. 97 shows a western blot of cell lysate from HT-29 cells treated with or without gamma interferon.
  • FIG. 98 shows a western blot of cell lysate from HT-29 cells that were transfected with either control siRNA or apoptosis-specific eIF-5A siRNAs. This figure shows that siRNAs of apoptosis-specific eIF-5A inhibit expression of apoptosis-specific eIF-5A.
  • FIG. 99 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs have a reduced level of TNF production.
  • FIG. 100 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs exhibit a decreased in apoptosis as compared to control cells. Both control and siRNA-transfected cells were primed with interferon gamma and also treated with TNF-α.
  • FIG. 101 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs express less TLR4 protein than control cells.
  • FIG. 102 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs express less TNFR1 protein than control cells.
  • FIG. 103 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs express less iNOS protein than control cells.
  • FIG. 104 shows that HT-29 cells transfected with apoptosis-specific eIF-5A siRNAs express less TLR4 mRNA than control cells.
  • FIG. 105 shows the time course for U937 treatments.
  • FIG. 106 shows that apoptosis-specific eIF-5A is upregulated with PMA in U937 cells.
  • FIG. 107 shows that apoptosis-specific eIF-5A is upregulated with LPS in U937 cells.
  • FIG. 108 shows that apoptosis-specific eIF-5A protein expression is still reduced after numerous hours following siRNA treatment.
  • FIG. 109 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction of TLR4.
  • FIG. 110 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with fewer glycosylated forms of the interferon gamma receptor in U937 cells.
  • FIG. 111 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction in TNFR1 in U937 cells.
  • FIG. 112 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction in LPS-induced TNF-α production in U937 cells.
  • FIG. 113 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction in LPS-induced IL-1β production in U937 cells.
  • FIG. 114 shows that siRNA mediated down-regulation of apoptosis-specific eIF-5A coincides with a reduction in LPS-induced IL-8 production in U937 cells.
  • FIG. 115 shows that IL-6 production is independent of siRNA mediated down-regulation of apoptosis-specific eIF-5A in U937 cells.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Several isoforms of eukaryotic initiation factor 5A (“eIF-5A”) have been isolated and present in published databanks. It was thought that these isoforms were functionally redundant. The present inventors have discovered that one isoform is upregulated immediately before the induction of apoptosis, which they have designated apoptosis-specific eIF-5A or eIF-5A1. The subject of the present invention is apoptosis-specific eIF-5A and DHS, which is involved in the activation of eIF-5A.
  • Apoptosis-specific eIF-5A is likely to be a suitable target for intervention in apoptosis-causing disease states since it appears to act at the level of post-transcriptional regulation of downstream effectors and transcription factors involved in the apoptotic pathway. Specifically, apoptosis-specific eIF-5A appears to selectively facilitate the translocation of mRNAs encoding downstream effectors and transcription factors of apoptosis from the nucleus to the cytoplasm, where they are subsequently translated. The ultimate decision to initiate apoptosis appears to stem from a complex interaction between internal and external pro- and anti-apoptotic signals. Lowe & Lin (2000) Carcinogenesis, 21, 485-495. Through its ability to facilitate the translation of downstream apoptosis effectors and transcription factors, apoptosis-specific eIF-5A appears to tip the balance between these signals in favor of apoptosis.
  • Accordingly, the present invention provides a method of suppressing or reducing apoptosis in a cell by administering an agent that inhibits or reduces expression of either apoptosis-specific eIF-5A or DHS. By reducing expression of DHS, there is less DHS protein to be available to activate apoptosis-specific eIF-5A. One agent that can inhibit or reduce expression of apoptosis-specific eIF-5A or DHS are antisense oligonucleotides of apoptosis-specific eIF-5A or DHS. By reducing activation of apoptosis-specific eIF-5A or by reducing or inhibiting expression of apoptosis-specific eIF-5A, cellular apoptosis can be delayed or inhibited.
  • Antisense oligonucleotides have been successfully used to accomplish both in vitro as well as in vivo gene-specific suppression. Antisense oligonucleotides are short, synthetic strands of DNA (or DNA analogs), RNA (or RNA analogs), or DNA/RNA hybrids that are antisense (or complimentary) to a specific DNA or RNA target. Antisense oligonucleotides are designed to block expression of the protein encoded by the DNA or RNA target by binding to the target mRNA and halting expression at the level of transcription, translation, or splicing. By using modified backbones that resist degradation (Blake et al., 1985), such as replacement of the phosphodiester bonds in the oligonucleotides with phosphorothioate linkages to retard nuclease degradation (Matzura and Eckstein, 1968), antisense oligonucleotides have been used successfully both in cell cultures and animal models of disease (Hogrefe, 1999). Other modifications to the antisense oligonucleotide to render the oligonucleotide more stable and resistant to degradation are known and understand by one skilled in the art. Antisense oligonucleotide as used herein encompasses double stranded or single stranded DNA, double stranded or single stranded RNA, DNA/RNA hybrids, DNA and RNA analogs, and oligonucleotides having base, sugar, or backbone modifications. The oligonucleotides may be modified by methods known in the art to increase stability, increase resistance to nuclease degradation or the like. These modifications are known in the art and include, but are not limited to modifying the backbone of the oligonucleotide, modifying the sugar moieties, or modifying the base.
  • Preferably, the antisense oligonucleotides of the present invention have a nucleotide sequence encoding a portion or the entire coding sequence of an apoptosis-specific eIF-5A polypeptide or a DHS polypeptide. The inventors have transfected various cell lines with antisense nucleotides encoding a portion of an apoptosis-specific eIF-5A polypeptide as described below and measured the number of cells undergoing apoptosis. The cell populations that were transfected with the antisense oligonucleotides showed a decrease in the number of cells undergoing apoptosis as compared to like cell populations not having been transfected with the antisense oligos. FIGS. 54-58 show a decrease in the percentage of cells undergoing apoptosis in the cells having being treated with antisense apoptosis-specific eIF-5A oligonucleotides as compared to cells not having been transfected with the antisense apoptosis-specific eIF-5A oligonucleotides.
  • The present invention contemplates the use of many suitable nucleic acid sequences encoding an apoptosis-specific eIF-5A polypeptide or DHS polypeptide. For example, the present invention provides antisense oligonucleotides of the following apoptosis-specific eIF-5A nucleic acid sequences (SEQ ID NOS: 1, 3, 4, 5, 11, 12, 15, 16, 19, 20, and 21) and DHS sequences (SEQ ID NOS:6, 7, 8). Antisense oligonucleotides of the present invention need not be the entire length of the provided SEQ ID NOs. They need only be long enough to be able to bind to the mRNA and inhibit expression of such mRNA. “Inhibition or reduction of expression” or “suppression of expression” refers to the absence or detectable decrease in the level of protein and/or mRNA product from a target gene, such as apoptosis-specific eIF-5A.
  • Exemplary antisense oligonucleotides of apoptosis-specific eIF-5A that do not comprise the entire coding sequence are antisense oligonucleotides of apoptosis-specific eIF-5A having the following SEQ ID NO: 35, 37, and 39.
  • “Antisense oligonucleotide of apoptosis-specific eIF-5A” includes oligonucleotides having substantial sequence identity or substantial homology to apoptosis-specific eIF-5A. Additional antisense oligonucleotides of apoptosis-specific eIF-5A of the present invention include those that have substantial sequence identity to those enumerated above (i.e. 90% homology) or those having sequences that hybridize under highly stringent conditions to the enumerated SEQ ID NOs. As used herein, the term “substantial sequence identity” or “substantial homology” is used to indicate that a sequence exhibits substantial structural or functional equivalence with another sequence. Any structural or functional differences between sequences having substantial sequence identity or substantial homology will be de minimus; that is, they will not affect the ability of the sequence to function as indicated in the desired application. Differences may be due to inherent variations in codon usage among different species, for example. Structural differences are considered de minimus if there is a significant amount of sequence overlap or similarity between two or more different sequences or if the different sequences exhibit similar physical characteristics even if the sequences differ in length or structure. Such characteristics include, for example, the ability to hybridize under defined conditions, or in the case of proteins, immunological crossreactivity, similar enzymatic activity, etc. The skilled practitioner can readily determine each of these characteristics by art known methods.
  • Additionally, two nucleotide sequences are “substantially complementary” if the sequences have at least about 70 percent or greater, more preferably 80 percent or greater, even more preferably about 90 percent or greater, and most preferably about 95 percent or greater sequence similarity between them. Two amino acid sequences are substantially homologous if they have at least 70%, more preferably at least 80%, even more preferably at least 90%, and most preferably at least 95% similarity between the active, or functionally relevant, portions of the polypeptides.
  • To determine the percent identity of two sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). In a preferred embodiment, at least 30%, 40%, 50%, 60%, 70%, 80%, or 90% or more of the length of a reference sequence is aligned for comparison purposes. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • The comparison of sequences and determination of percent identity and similarity between two sequences can be accomplished using a mathematical algorithm. (Computatio nal Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991).
  • The nucleic acid and protein sequences of the present invention can further be used as a “query sequence” to perform a search against sequence databases to, for example, identify other family members or related sequences. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol Biol. 215:403-10. BLAST nucleotide searches can be performed with the NBLAST program. BLAST protein searches can be performed with the XBLAST program to obtain amino acid sequences homologous to the proteins of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25(17):3389-3402. When utilizing BLAST and gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used.
  • The term “apoptosis-specific eIF-5A” includes functional derivatives thereof. The term “functional derivative” of a nucleic acid is used herein to mean a homolog or analog of the gene or nucleotide sequence. A functional derivative may retain the function of the given gene, which permits its utility in accordance with the invention. “Functional derivatives” of the apoptosis-specific eIF-5A polypeptide or functional derivatives of antisense oligonucleotides of apoptosis-specific eIF-5A as described herein are fragments, variants, analogs, or chemical derivatives of apoptosis-specific eIF-5A that retain Apoptosis-specific eIF-5A activity or immunological cross reactivity with an antibody specific for apoptosis-specific eIF-5A. A fragment of the apoptosis-specific eIF-5A polypeptide refers to any subset of the molecule.
  • Functional variants can also contain substitutions of similar amino acids that result in no change or an insignificant change in function. Amino acids that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham et al. (1989) Science 244:1081-1085). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity such as kinase activity or in assays such as an in vitro proliferative activity. Sites that are critical for binding partner/substrate binding can also be determined by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling (Smith et al. (1992) J. Mol. Biol. 224:899-904; de Vos et al. (1992) Science 255:306-312).
  • A “variant” refers to a molecule substantially similar to either the entire gene or a fragment thereof, such as a nucleotide substitution variant having one or more substituted nucleotides, but which maintains the ability to hybridize with the particular gene or to encode mRNA transcript which hybridizes with the native DNA. A “homolog” refers to a fragment or variant sequence from a different animal genus or species. An “analog” refers to a non-natural molecule substantially similar to or functioning in relation to the entire molecule, a variant or a fragment thereof.
  • Variant peptides include naturally occurring variants as well as those manufactured by methods well known in the art. Such variants can readily be identified/made using molecular techniques and the sequence information disclosed herein. Further, such variants can readily be distinguished from other proteins based on sequence and/or structural homology to the eIF-5A or DHS proteins of the present invention. The degree of homology/identity present will be based primarily on whether the protein is a functional variant or non-functional variant, the amount of divergence present in the paralog family and the evolutionary distance between the orthologs.
  • Non-naturally occurring variants of the eIF-5A or DHS polynucleotides, antisense oligonucleotides, or proteins of the present invention can readily be generated using recombinant techniques. Such variants include, but are not limited to deletions, additions and substitutions in the nucleotide or amino acid sequence. For example, one class of substitutions are conserved amino acid substitutions. Such substitutions are those that substitute a given amino acid in a protein by another amino acid of like characteristics. Typically seen as conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu, and Ile; interchange of the hydroxyl residues Ser and Thr; exchange of the acidic residues Asp and Glu; substitution between the amide residues Asn and Gln; exchange of the basic residues Lys and Arg; and replacements among the aromatic residues Phe and Tyr. Guidance concerning which amino acid changes are likely to be phenotypically silent are found in Bowie et al., Science 247:1306-1310 (1990).
  • The term “hybridization” as used herein is generally used to mean hybridization of nucleic acids at appropriate conditions of stringency as would be readily evident to those skilled in the art depending upon the nature of the probe sequence and target sequences. Conditions of hybridization and washing are well known in the art, and the adjustment of conditions depending upon the desired stringency by varying incubation time, temperature and/or ionic strength of the solution are readily accomplished. See, e.g. Sambrook, J. et al., Molecular Cloning: A Laboratory Manual, 2nd edition, Cold Spring Harbour Press, Cold Spring Harbor, N.Y., 1989.
  • The choice of conditions is dictated by the length of the sequences being hybridized, in particular, the length of the probe sequence, the relative G-C content of the nucleic acids and the amount of mismatches to be permitted. Low stringency conditions are preferred when partial hybridization between strands that have lesser degrees of complementarity is desired. When perfect or near perfect complementarity is desired, high stringency conditions are preferred. High stringency conditions means that the hybridization solution contains 6×S.S.C., 0.01 M EDTA, 1× Denhardt's solution and 0.5% SDS. Hybridization is carried out at about 68° C. for about 3 to 4 hours for fragments of cloned DNA and for about 12 to 16 hours for total eucaryotic DNA. For lower stringencies, the temperature of hybridization is reduced to about 42° C. below the melting temperature (Tm) of the duplex. The Tm is known to be a function of the G-C content and duplex length as well as the ionic strength of the solution.
  • As used herein, the phrase “hybridizes to a corresponding portion” of a DNA or RNA molecule means that the molecule that hybridizes, e.g., oligonucleotide, polynucleotide, or any nucleotide sequence (in sense or antisense orientation) recognizes and hybridizes to a sequence in another nucleic acid molecule that is of approximately the same size and has enough sequence similarity thereto to effect hybridization under appropriate conditions. For example, a 100 nucleotide long sense molecule will recognize and hybridize to an approximately 100 nucleotide portion of a nucleotide sequence, so long as there is about 70% or more sequence similarity between the two sequences. It is to be understood that the size of the “corresponding portion” will allow for some mismatches in hybridization such that the “corresponding portion” may be smaller or larger than the molecule which hybridizes to it, for example 20-30% larger or smaller, preferably no more than about 12-15% larger or smaller.
  • In addition, functional variants of polypeptides can also contain substitution of similar amino acids that result in no change or an insignificant change in function. Amino acids that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham et al., Science 244:1081-1085 (1989)). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity or in assays.
  • The present invention also provides other agents that can inhibit or reduce expression of apoptosis-specific eIF-5A or DHS. One such agent includes small inhibitory RNAs (“siRNA”). siRNA technology has been emerging as a viable alternative to antisense oligonucleotides since lower concentrations are required to achieve levels of suppression that are equivalent or superior to those achieved with antisense oligonucleotides (Thompson, 2002). Long double-stranded RNAs have been used to silence the expression of specific genes in a variety of organisms such as plants, nematodes, and fruit flies. An RNase-III family enzyme called Dicer processes these long double stranded RNAs into 21-23 nucleotide small interfering RNAs which are then incorporated into an RNA-induced silencing complex (RISC). Unwinding of the siRNA activates RISC and allows the single-stranded siRNA to guide the complex to the endogenous mRNA by base pairing. Recognition of the endogenous mRNA by RISC results in its cleavage and consequently makes it unavailable for translation. Introduction of long double stranded RNA into mammalian cells results in a potent antiviral response, which can be bypassed by use of siRNAs. (Elbashir et al., 2001). siRNA has been widely used in cell cultures and routinely achieves a reduction in specific gene expression of 90% or more.
  • The use of siRNAs has also been gaining popularity in inhibiting gene expression in animal models of disease. A recent study demonstrated that an siRNA against luciferase was able to block luciferase expression from a co-transfected plasmid in a wide variety of organs in post-natal mice. (Lewis et al., 2002). An siRNA against Fas, a receptor in the TNF family, injected hydrodynamically into the tail vein of mice was able to transfect greater than 80% of hepatocytes and decrease Fas expression in the liver by 90% for up to 10 days after the last injection (Song et al., 2003). The Fas siRNA was also able to protect mice from liver fibrosis and fulminant hepatitis. The development of sepsis in mice treated with a lethal dose of lipopolysaccharide was inhibited by the use of an siRNA directed against TNF-α (Sorensen et al., 2003). SiRNA has the potential to be a very potent drug for the inhibition of specific gene expression in vitro in light of their long-lasting effectiveness in cell cultures and their ability to transfect cells in vivo and their resistance to degradation in serum in vivo (Bertrand et al., 2002) in vivo.
  • The present inventors have transfected cells with siRNAs of apoptosis-specific eIF-5A and studied the effects on expression of apoptosis-specific eIF-5A. FIG. 64 shows that cells transfected with apoptosis-specific eIF-5A siRNA produced less apoptosis-specific eIF-5A protein. FIGS. 65-67 show that cell populations transfected with apoptosis-specific eIF-5A siRNAs have a lower percentage of cells undergoing apoptosis after exposure to amptothecin and TNF-α as compared to cells not having been transfected with apoptosis-specific eIF-5A siRNAs. Thus, one embodiment of the present invention provides for inhibiting expression of apoptosis-specific eIF-5A in cells by transfecting the cells with a vector comprising a siRNAs of apoptosis-specific eIF-5A.
  • Preferred siRNAs of apoptosis-specific eIF-5A include those that have SEQ ID NO: 31, 31, 32, and 33. Additional siRNAs include those that have substantial sequence identity to those enumerated (i.e. 90% homology) or those having sequences that hybridize under highly stringent conditions to the enumerated SEQ ID NOs. What is meant by substantial sequence identity and homology is described above with respect to antisense oligonucleotides of the present invention. The term “siRNAs of apoptosis-specific eIF-5A” include functional variants or derivatives as described above with respect to antisense oligonucleotides of the present invention.
  • Delivery of siRNA and expression constructs/vectors comprising siRNA are known by those skilled in the art. U.S. applications 2004/106567 and 2004/0086884, which are herein incorporated by reference in their entirety, provide numerous expression constructs/vectors as well as delivery mechanism including viral vectors, non viral vectors, liposomal delivery vehicles, plasmid injection systems, artificial viral envelopes and poly-lysine conjugates to name a few.
  • One skilled in the art would understand regulatory sequences useful in expression constructs/vectors with antisense oligonucleotides or siRNA. For example, regulatory sequences may be a constitutive promoter, an inducible promoter, a tissue-specific promoter, or a combination thereof.
  • Many important human diseases are caused by abnormalities in the control of apoptosis. These abnormalities can result in either a pathological increase in cell number (e.g. cancer) or a damaging loss of cells (e.g. degenerative diseases). As non-limiting examples, the methods and compositions of the present invention can be used to prevent or treat the following apoptosis-associated diseases and disorders: neurological/ neurodegenerative disorders (e.g., Alzheimer's, Parkinson's, Huntington's, Amyotrophic Lateral Sclerosis (Lou Gehrig's Disease), autoimmune disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus (SLE), multiple sclerosis), Duchenne Muscular Dystrophy (DMD), motor neuron disorders, ischemia, heart ischemia, chronic heart failure, stroke, infantile spinal muscular atrophy, cardiac arrest, renal failure, atopic dermatitis, sepsis and septic shock, AIDS, hepatitis, glaucoma, diabetes (type 1and type 2), asthma, retinitis pigmentosa, osteoporosis, xenograft rejection, and burn injury.
  • One such disease caused by abnormalities in the control of apoptosis is glaucoma. Apoptosis in various optical tissues is a critical factor-leading to blindness in glaucoma patients. Glaucoma is a group of eye conditions arising from damage to the optic nerve that results in progressive blindness. Apoptosis has been shown to be a direct cause of this optic nerve damage.
  • Early work in the field of glaucoma research has indicated that elevated intra-ocular pressure (“IOP”) leads to interference in axonal transport at the level of the lamina cribosa (a perforated, collagenous connective tissue) that is followed by the death of retinal ganglion cells. Quigley and Anderson (1976) Invest. Ophthalmol. Vis. Sci., 15, 606-16; Minckler, Bunt, and Klock, (1978) Invest. Ophthalmol. Vis. Sci., 17, 33-50; Anderson and Hendrickson, (1974) Invest. Ophthalmol. Vis. Sci., 13, 771-83; Quigley et al., (1980) Invest. Ophthalmol. Vis. Sci., 19, 505-17. Studies of animal models of glaucoma and post-mortem human tissues indicate that the death of retinal ganglion cells in glaucoma occurs by apoptosis. Garcia-Valenzuela e. al., (1995) Exp. Eye Res., 61, 33-44; Quigley et al., (1995) Invest. Ophthalmol. Vis. Sci., 36, 774-786; Monard, (1998) In: Haefliger IO, Flammer J (eds) Nitric Oxide and Endothelin in the Pathogenesis of Glaucoma, New York, N.Y., Lippincott-Raven, 213-220. The interruption of axonal transport as a result of increased IOP may contribute to retinal ganglion cell death by deprivationoftrophic factors. Quigley, (1995) Aust N Z J Ophthalmol, 23(2), 85-91. Optic nerve head astrocytes in glaucomatous eyes have also been found to produce increased levels of some neurotoxic substances. For example, increased production of tumor necrosis factor-α (TNF-α) (Yan et al., (2000) Arch. Ophthalmol., 118, 666-673), and nitric oxide synthase (Neufeld et al., (1997) Arch. Ophthalmol., 115, 497-503), the enzyme which gives rise to nitric oxide, has been found in the optic nerve head of glaucomatous eyes. Furthermore, increased expression of the inducible form of nitric oxide synthase (iNOS) and TNF-α by activated retinal glial cells have been observed in rat models of hereditary retinal diseases. Cotinet et al., (1997) Glia, 20, 59-69; de Kozak et al., (1997) Ocul. Immunol. Inflamm., 5, 85-94; Goureau et al., (1999) J. Neurochem, 72, 2506-2515. In the glaucomatous optic nerve head, excessive nitric oxide has been linked to the degeneration of axons of retinal ganglion cells. Arthur and Neufeld, (1999) Surv Ophthalmol, 43 (Suppl 1), S129-S135. Finally, increased production of TNF-α by retinal glial cells in response to simulated ischemia or elevated hydrostatic pressure has been shown to induce apoptosis in co-cultured retinal ganglion cells. Tezel and Wax, (2000) J. Neurosci., 20(23), 8693-8700.
  • Protecting retinal ganglion cells from degeneration by apoptosis is under study as a potential new treatment for blindness due to glaucoma. Antisense oligonucleotides have been used successfully in animal models of eye disease. In a model of transient global retinal ischemia, expression of caspase 2 was increased during ischemia, primarily in the inner nuclear and ganglion cell layers of the retina. Suppression of caspase using an antisense oligonucleotide led to significant histopathologic and functional improvement as determined by electroretinogram. Singh et al., (2001) J. Neurochem., 77(2), 466-75. Another study demonstrated that, upon transfection of the optic nerve, retinal ganglion cells upregulate the pro-apoptotic protein Bax and undergo apoptosis. Repeated injections of a Bax antisense oligonucleotide into the temporal superior retina of rats inhibited the local expression of Bax and increased the number of surviving retinal ganglion cells following transaction of the optic nerve. Isenmann et al., (1999) Cell Death Differ., 6(7). 673-82.
  • Delivery of antisense oligonucleotides to retinal ganglion cells has been improved by encapsulating the oligonucleotides in liposomes, which were then coated with the envelope of inactivated hemagglutinating virus of Japan (HVJ; Sendai virus) by fusion (HVJ liposomes). Intravitreal injection into mice of FITC-labeled antisense oligonucleotides encapsulated in HVJ liposomes resulted in high fluorescence within 44% of the cells in the ganglion layer which lasted three days while fluorescence with naked FITC-labeled antisense oligonucleotide disappeared after one day. Hangai et al., (1998) Arch Ophthalmol, 116(7), 976.
  • One method of preventing or reducing apoptosis of the present invention is directed to preventing or reducing apoptosis in cells and tissues of the eye, such as but not limited to, astrocytes, retinal ganglion, retinal glial cells and lamina cribosa. Death of retinal ganglion cells in glaucoma occurs by apoptosis and which leads to blindness. Thus, providing a method of inhibiting or reducing apoptosis in retinal ganglion cells or by protecting retinal ganglion cells from degeneration by apoptosis provides a novel treatment for prevention of blindness due to glaucoma.
  • The present invention provides a method for preventing or inhibiting retinal ganglion cell death in a glaucomatous eye, by suppressing expression of apoptosis-specific eIF-5A. Inhibiting the expression of apoptosis-specific eIF-5A reduces apoptosis. Apoptosis-specific eIF-5A is a powerful gene that appears to regulate the entire apoptotic process. Thus, controlling apoptosis in the optic nerve head indicates that blocking expression of apoptosis-specific eIF-5A provides a treatment for glaucoma.
  • Suppression of expression of apoptosis-specific eIF-5A is accomplished by administering an antisense oligonucleotides or a siRNA of human apoptosis-specific eIF-5A to cells of the eye such as, but not limited to lamina cribrosa, astrocytes, retinal ganglion, or retinal glial cells. Antisense oligonucleotides and siRNAs are as defined above, i.e. have a nucleotide sequence encoding at least a portion of an apoptosis-specific eIF-5A polypeptide. Exemplary antisense oligonucleotides useful in this aspect of the invention comprise SEQ ID NO:26 or 27 or oligonucleotides that bind to a sequence complementary to SEQ ID NO:26 or 27 under high stringency conditions and which inhibit expression of apoptosis-specific eIF-5A.
  • Another embodiment of the invention provides a method of suppressing expression of apoptosis-specific eIF-5A in lamina cribosa cells, astrocyte cells, retinal ganglion cells or retinal glial cells. Antisense oligonucleotides or siRNAs, such as but not limited to, SEQ ID NO:26 and 27, targeted against human apoptosis-specific eIF-5A are administered to lamina cribosa cells, astrocyte cells, retinal ganglion cells or retinal glial cells. The cells may be of human origin.
  • In addition to having a role in apoptosis, eIF5A may also play a role in the immune response. The present inventors have discovered that apoptosis-specific eIF-5A levels correlate with elevated levels of two cytokines (Interleukin 1-beta “IL-1β” and interleukin 18 “IL-18”) in ischemic heart tissue, thus further proving that apoptosis-specific eIF-5A is involved in cell death as it is present in ischemic heart tissue. This apoptosis-specific eIF-5A/interleukin correlation is not seen in non-ischemic heart tissue. See FIGS. 50A-F and 51. Using PCR measurements, levels of apoptosis-specific eIF-5A, and proliferating eIF-5A (“e lF-5A2”)—another isoform), IL-1β, and IL-18 were measured and compared in various ischemic heart tissue (from coronary bypass graft and valve (mitral and atrial valve) replacement patients).
  • The correlation of apoptosis-specific eIF-5A to these potent interleukins further suggests that the inflammation and apoptosis pathways in ischemia may be controlled via controlling levels of apoptosis-specific eIF-5A. Further evidence that apoptosis-specific eIF-5A is involved in the immune response is suggested by the fact that human peripheral blood mononuclear cells (PBMCs) normally express very low levels of eIF-5A, but upon stimulation with T-lymphocyte-specific stimuli expression of apoptosis-specific eIF-5A increases dramatically (Bevec et al., 1994). This suggests a role for apoptosis-specific eIF-5A in T-cell proliferation and/or activation. Since activated T cells are capable of producing a wide variety of cytokines, it is also possible that apoptosis-specific eIF-5A may be required as a nucleocytoplasmic shuttle for cytokine mRNAs. The authors of the above referenced article also found elevated levels of eIF5A in the PBMCs of HIV-1 patients which may contribute to efficient HIV replication in these cells as eIF5A has been demonstrated to be a cellular binding factor for the HIV Rev protein and required for HIV replication (Ruhl et al., 1993).
  • More recently, eIF-5A expression was found to be elevated during dendritic cell maturation (Kruse et al, 2000). Dendritic cells are antigen-presenting cells that sensitize helper and killer T cells to induce T cell-mediated immunity (Steinman, 1991). Immature dendritic cells lack the ability to stimulate T cells and require appropriate stimuli (i.e. inflammatory cytokines and/or microbial products) to mature into cells capable of activating T cells. An inhibitor of deoxyhypusine synthase, the enzyme required to activate eIF5A, was found to inhibit T lymphocyte activation by dendritic cells by preventing CD83 surface expression (Kruse et aL, 2000). Thus, eIF5A may facilitate dendritic cell maturation by acting as a nucleocytoplasmic shuttle for CD83 mRNA.
  • In both of these studies (Bevec et al., 1994; Kruse et al., 2000) implicating a role for eIF5A in the immune system, the authors did not specify nor identify which isoform of eIF5A they were examining, nor did they have a reason to. As discussed above, humans are known to have two isoforms of eIF5A, apoptosis-specific eIF-5A (“eIF5A1”) and proliferating eIF-5A (“eIF-5A2”), both encoded on separate chromosomes. Prior to the present inventors discoveries it was believed that both of these isoforms were functional redundant. The oligonucleotide described by Bevec et al. that was used to detect eIF5A mRNA in stimulated PBMCs had 100% homology to human apoptosis-specific eIF-5A and the study pre-dates the cloning of proliferating eIF-5A. Similarly, the primers described by Kruse et al. that were used to detect eIEF5A by reverse transcription polymerase chain reaction during dendritic cell maturation had 100% homology to human apoptosis-specific eIF-5A.
  • The present invention relates to controlling the expression of apoptosis-specific eIF-5A to control the rate of dendritic cell maturation and PBMC activation, which in turn may control the rate of T cell-mediated immunity. The present inventors studied the role of apoptosis-specific eIF-5A in the differentiation of monocytes into adherent macrophages using the U-937 cell line, as U-937 is known to express eIF-5A mRNA (Bevec et al., 1994). U-937 is a human monocyte cell line that grows in suspension and will become adherent and differentiate into macrophages upon stimulation with PMA. When PMA is removed by changing the media, the cells become quiescent and are then capable of producing cytokines (Barrios-Rodiles et al., J. Immunol., 163:963-969 (1999)). In response to lipopolysaccharide (LPS), a factor found on the outer membrane of many bacteria known to induce a general inflammatory response, the macrophages produce both TNF-α and IL-1β (Barrios-Rodiles et al., 1999). See FIG. 78 showing a chart of stem cell differentiation and the resultant production of cytokines. The U-937 cells also produce IL-6 and IL-10 following LPS-stimulation (Izeboud et al., J. Receptor & Signal Transduction Research, 19(1-4):191-202. (1999)).
  • Using U-937 cells, it was shown that apoptosis-specific eIF-5A is upregulated during monocyte differentiation and TNF-α secretion. See FIG. 77. Accordingly, one aspect of the invention provides for a method of inhibiting or delaying maturation of macrophages to inhibit or reduce the production of cytokines. This method involves providing an agent that is capable of reducing the expression of either DHS or apoptosis-specific eIF-5A. By reducing or eliminating expression of DHS, apoptosis-specific eIF-5A activation will be reduced or eliminated. Since, apoptosis-specific eIF-5A is upregulated during monocyte differentiation and TNF-α secretion, it is believed that it is necessary for these events to occur. Thus, by reducing or eliminating activation of apoptosis-specific eIF-5A or by directly reducing or eliminating apoptosis-specific eIF-5A expression, monocyte differentiation and TNF-α secretion can be reduced or eliminated. Any agent capable of reducing the expression of DHS or apoptosis-specific eIF-5A may be used and includes, but is not limited to antisense oligonucleotides or siRNAs as described herein.
  • The present inventors have studied the ability of human apoptosis-specific eIF-5A to promote translation of cytokines by acting as a nucleocytoplasmic shuttle for cytokine mRNAs in vitro using a cell line known to predictably produce cytokine(s) in response to a specific stimulus. Some recent studies have found that human liver cell lines can respond to cytokine stimulation by inducing production of other cytokines. HepG2 is a well characterized human hepatocellular carcinoma cell line found to be sensitive to cytokines. In response to IL-1β, HepG2 cells rapidly produce TNF-α mRNA and protein in a dose-dependent manner (Frede et al., 1996; Rowell et al., 1997; Wordemann et al., 1998). Thus, HepG2 cells were used as a model system to study the regulation of TNF-α production. The present inventors have shown that inhibition of human apoptosis-specific eIF-5A expression in HepG2 cells caused the cells to produce less TNF-α after having been transfected with antisense oligonucleotide of directed toward apoptosis factor 5A.
  • Thus one embodiment of the present invention provides a method for reducing levels of a cytokine. The method involves administering an agent capable of reducing expression of apoptosis factor 5A1. Reducing expression of apoptosis factor 5A1 also reduces expression of the cytokine and thus leads to a decreased amount of the cytokine produced by cell. The cytokine is a preferably a pro-inflammatory cytokine, including, but not limited to IL-1, IL-18, IL-6 and TNF-α.
  • Antisense oligonucleotides are as discussed above. Exemplary antisense oligonucleotides of human apoptosis-specific eIF-5A are selected from the group consisting of SEQ ID NO: 35, 37, and 39 or is an antisense nucleotide that hybridizes under highly stringent conditions to a sequence selected from the group consisting of SEQ ID NO: 35, 37, and 39.
  • An agent may also comprise a siRNA of human apoptosis-specific eIF-5A and are as discussed above. Exemplary siRNAs have a sequence selected from the group consisting of SEQ ID NO: 30, 31, 32 and 33 or is a siRNA that hybridizes under highly stringent conditions to a sequence selected from the group consisting of SEQ ID NO: 30, 31, 32 and 33. FIGS. 65-67 show that cells transfected with human apoptosis-specific eIF-5A siRNAs have a lower percentage of cells undergoing apoptosis after exposure to amptothecin and TNF-α.
  • The present invention is also directed to a method for reducing the expression of p53. This method involves administering an agent capable of reducing expression of apoptosis factor 5A, such as the antisense oligonucleotides or the siRNAs described above. Reducing expression of apoptosis-specific eIF-5A reduces expression of p53 as shown in FIG. 52 and example 10.
  • The present invention is also directed to a method for increasing the expression of Bcl-2. This method entails administering an agent capable of reducing expression of human apoptosis-specific eIF-5A. Preferred agents include antisense oligonucleotides and siRNAs described above. Reducing expression of apoptosis-specific eIF-5A increases expression of Bcl-2 as shown in FIG. 63 and example 13. FIG. 63 shows that cells transfected with apoptosis-specific eIF-5A siRNA produced less apoptosis-specific eIF-5A protein and in addition, produced more Bcl-2 protein. A decrease in apoptosis-specific eIF-5A expression correlates with an increase in BCL-2 expression.
  • The present invention also provides a method for reducing levels of TNF-alpha in a patient in need thereof comprising administering to said patient either antisense oligonucleotide or siRNAs of apoptosis-specific eIF-5A as described above. As demonstrated in FIG. 69 and example 14, cells transfected with antisense apoptosis-specific eIF-5A oligonucleotides of the present invention produced less TNF-α after induction with IL-1than cells not transfected with such antisense oligonucleotides.
  • Further, the present invention provides a method of treating pathological conditions characterized by an increased IL-1, TNF-alpha, IL-61 or IL-18 level comprising administering to a mammal having said pathological condition, agents to reduce expression of apoptosis-specific eIF-5A as described above (antisense oligonucleotides and siRNA).
  • Known pathological conditions characterized by an increase in IL-1, TNF-alpha, or Il-6 levels include, but are not limited to arthritis-rheumatoid and osteo arthritis, asthma, allergies, arterial inflammation, crohn's disease, inflammatory bowel disease, (ibd), ulcerative colitis, coronary heart disease, cystic fibrosis, diabetes, lupus, multiple sclerosis, graves disease, periodontitis, glaucoma & macular degeneration, ocular surface diseases including keratoconus, organ ischemia-heart, kidney, repurfusion injury, sepsis, multiple myeloma, organ transplant rejection, psoriasis and eczema. Thus, reducing expression of apoptosis-specific eIF-5A with the antisense oligonucleotids, siRNAs and methods of the present invention, may provide relief from these pathological conditions.
  • The present invention also provides a method of delivering siRNA to mammalian lung cells in vivo. siRNAs directed against apoptosis-specific eIF-5A were administered intranasally (mixed with water) to mice. 24 hours after administration of the siRNA against apoptosis-specific eIF-5A, lipopolysaccharide (LPS) was administered intranasally to the mice. After another 24 hours, the right lung was removed and myeloperoxidase was measured. The mouse apoptosis-specific eIF-5A siRNA suppressed myeloperoxidase by nearly 90% as compared to the control siRNA. In the study, there were 5 mice in each group. The results of this study show that siRNA can be delivered successfully in vivo to lung tissue in mammals, and that siRNA directed against apoptosis-specific eIF-5A inhibits the expression of apoptosis-specific eIF-5A resulting in a suppression of myeloperoxidase production. Myeloperoxidase (“MPO”) is a lysosomal enzyme that is found in neutrophils. The myeloperoxidase is an enzyme that uses hydrogen peroxidase to convert chloride to hypochlorous acid. The hypochlorous acid reacts with and destroys bacteria. Myeloperoxidase is also produced when arteries are inflamed. Thus, it is clear that myeloperoxidase is associated with neutrophils and the inflammation response. The present inventors have shown that by down regulating apoptosis-specific eIF-5A with siRNAs shows a marked decrease in the myeloperoxidase in lung tissue after exposed to LPS (which normally produces an inflarnmatory response involving the production of myeloperoxidase). Thus, the present inventors have shown that using siRNAs against apoptosis-specific eIF-5A can decrease or suppress the amount of myeloperoxidase in lung tissue and thus decrease or suppress the inflammation response.
  • LPS is a macromolecular cell surface antigen of bacteria that when applied in vivo triggers a network of inflammatory responses. Intranasally delivering LPS causes an increase in the number of neutrophils in the lungs. One of the primary events is the activation of mononuclear phagocytes through a receptor-mediated process, leading to the release of a number of cytokines, including TNF-α. In turn, the increased adherence of neutrophils to endothelial cells induced by TNF-α leads to massive infiltration in the pulmonary space.
  • Thus, not only have the present inventors shown the correlation between apoptosis-specific eIF-5A and the immune response, as well as shown that siRNAs against apoptosis-specific eIF-5A an suppress the production of myeloperoxidase (i.e. part of the inflammation response). The inventors have also shown that it is possible to deliver siRNAs in vivo to lung tissue by simple intranasal delivery. The siRNAs were mixed only in water. This presents a major breakthrough and discovery as others skilled in the art have attempted to design acceptable delivery methods for siRNA.
  • The ability to reduce inflammation is of direct importance in many diseases. MPO levels are a critical predictor of heart attacks and cytokine-induced inflammation caused by autoimmune disorders. This ability to decrease or suppress the inflammation response may serve useful in treating inflammation related disorders such as auto immune disorders. In addition, the ability to lower MPO could be a means of protecting patients from ischemic events and heart attacks.
  • In another experiment, mice were similarly treated with siRNAs directed against apoptosis-specific eIF-5A. Lipopolysaccharide (LPS) was administered to the mice to induce inflammation and an immune system response. Under control conditions, LPS kills thymocytes, which are important immune system precursor cells created in the thymus to fend off infection. However, using the siRNAs directed against apoptosis-specific eIF-5A allowed approximately 90% survivability of the thymocytes in the presence of LPS. When thymocytes are destroyed, since they are precursors to T cells, the body's natural immunity is compromised by not being able to produce T cells and thus can't ward off bacterial infections and such. Thus, using the siRNAs against apoptosis-specific eIF-5A can be used to reduce inflammation (as shown by a lower level of MPO in the first example) without destroying the body's natural immune defense system.
  • One embodiment of the present invention relates to reducing NFk beta (“NFkB”) levels by inhibiting apoptosis-specific eIF-5A with siRNAs targeted at apoptosis-specific eIF-5A. NFk beta is a major cell-signaling molecule for inflammation—its activation induces the expression of COX-2, which leads to tissue inflammation. The expression of the COX-2-encoding gene, believed to be responsible for the massive production of prostaglandins at inflammatory sites, is transcriptionaly regulated by NFkB. NFkB resides in the cytoplasm of the cell and is bound to its inhibitor. Injurious and inflammatory stimuli release NFkB from the inhibitor. NFkB moves into the nucleus and activates the genes responsible for expressing COX-2. Thus, by reducing levels of NFk beta, inflammation can be reduced.
  • In one experiment human epithelial cells (HT-29 cells) were treated with siRNA targeted at apoptosis-specific eIF-5A. Inflammation was then induced by NFkB by addition of TNF or interferon gamma and LPS for one hour. The results of this experiment show that inhibiting the expression of apoptosis-specific eIF-5A with siRNAs provided for a reduction in the levels of NFKB that were activated by the gamma interferon and LPS. See FIG. 74.
  • One embodiment of the present invention provides methods of inhibition expression of endogenous apoptosis-specific eIF-5A in a cell. Inhibiting expression is preferably carried out by the use of antisense polynucleotides or siRNAs of apoptosis-specific eIF-5A of the present invention described previously. When expression of endogenous apoptosis-specific eIF-5A occurs various effects on the cell result. For example, a reduction in expression of the various proteins, factors, receptors, cytokines are seen: p53; pro-inflammatory cytokines (See FIG. 112, 113, 114); myeloperoxidase; active NFk beta; TLR4 (See FIG. 109); TNFR-1 (See FIG. 111) and iNOS. A reduction in expression of endogenous apoptosis-specific eIF-5A increases levels expression of Bcl-2 in the cell.
  • It is understood that the antisense nucleic acids siRNAs of the present invention, where used in an animal for the purpose of prophylaxis or treatment, will be administered in the form of a composition additionally comprising a pharmaceutically acceptable carrier. Suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. Pharmaceutically acceptable carriers can further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the binding proteins. The compositions of the injection can, as is well known in the art, be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the mammal.
  • The compositions of this invention can be in a variety of forms. These include, for example, solid, semi-solid and liquid dosage forms, such as tablets, pills, powders, liquid solutions, dispersions or suspensions, liposomes, suppositories, injectable and infusible solutions. The preferred form depends on the intended mode of administration and therapeutic application.
  • Such compositions can be prepared in a manner well known in the pharmaceutical art. In making the composition the active ingredient will usually be mixed with a carrier, or diluted by a carrier, and/or enclosed within a carrier which can, for example, be in the form of a capsule, sachet, paper or other container. When the carrier serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, excipient or medium for the active ingredient. Thus, the composition can be in the form of tablets, lozenges, sachets, cachets, elixirs, suspensions, aerosols (as a solid or in a liquid medium), ointments containing for example up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, injection solutions, suspensions, sterile packaged powders and as a topical patch.
  • Having now generally described the invention, the same will be more readily understood through reference to the following examples, which are provided by way of illustration. The Examples are set forth to aid in understanding the invention but are not intended to, and should not be construed to, limit its scope in any way. The examples do not include detailed descriptions of conventional methods. Such methods are well known to those of ordinary skill in the art and are described in numerous publications. Detailed descriptions of conventional methods, such as those employed in the construction of vectors and plasmids, the insertion of nucleic acids encoding polypeptides into such vectors and plasmids, the introduction of plasmids into host cells, and the expression and determination thereof of genes and gene products can be obtained from numerous publication, including Sambrook, J. et al., (1989) Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press. All references mentioned herein are incorporated in their entirety.
  • EXAMPLES Example 1
  • Visualization of Apoptosis in Rat Corpus Luteum by DNA Laddering
  • The degree of apoptosis was determined by DNA laddering. Genomic DNA was isolated from dispersed corpus luteal cells or from excised corpus luteum tissue using the QIAamp DNA Blood Kit (Qiagen) according to the manufacturer's instructions. Corpus luteum tissue was excised before the induction of apoptosis by treatment with PGF-2α, 1 hour and 24 hours after induction of apoptosis. The isolated DNA was end-labeled by incubating 500 ng of DNA with 0.2 μCi[α-32P]dCTP, 1 mM Tris, 0.5 mM EDTA, 3 units of Klenow enzyme, and 0.2 pM each of dATP, dGTP, and dTTP at room temperature for 30 minutes. Unincorporated nucleotides were removed by passing the sample through a 1 ml Sepadex G-50 column according to Sambrook et al. The samples were then resolved by Tris-acetate-EDTA (1.8%) gel electrophoresis. The gel was dried for 30 minutes at room temperature under vacuum and exposed to x-ray film at −80° C. for 24 hours.
  • In one experiment, the degree of apoptosis in superovulated rat corpus lutea was examined either 0, 1, or 24 hours after injection with PGF-2α. In the 0 hour control, the ovaries were removed without PGF-2αinjection. Laddering of low molecular weight DNA fragments reflecting nuclease activity associated with apoptosis is not evident in control corpus luteum tissue excised before treatment with PGF-2α, but is discernible within 1 hour after induction of apoptosis and is pronounced by 24 hours after induction of apoptosis, which is shown in FIG. 16. In this figure, the top panel is an autoradiograph of the Northern blot probed with the 32P-dCTP-labeled 3′-untranslated region of rat corpus luteum apoptosis-specific DHS cDNA. The lower panel is the ethidium bromide stained gel of total RNA. Each lane contains 10 μg RNA. The data indicate that there is down-regulation of apoptosis-specific eIF-5A transcript following serum withdrawal.
  • In another experiment, the corresponding control animals were treated with saline instead of PGF-2α. Fifteen minutes after treatment with saline or PGF-2α, corpora lutea were removed from the animals. Genomic DNA was isolated from the corpora lutea at 3 hours and 6 hours after removal of the tissue from the animals. DNA laddering and increased end labeling of genomic DNA are evident 6 hours after removal of the tissue from the PGF-2α-treated animals, but not at 3 hours after removal of the tissue. See FIG. 17. DNA laddering reflecting apoptosis is also evident when corpora lutea are excised 15 minutes after treatment with PGF-2α and maintained for 6 hours under in vitro conditions in EBSS (Gibco). Nuclease activity associated with apoptosis is also evident from more extensive end labeling of genomic DNA.
  • In another experiment, superovulation was induced by subcutaneous injection with 500 μg of PGF-2α. Control rats were treated with an equivalent volume of saline solution. Fifteen to thirty minutes later, the ovaries were removed and minced with collagenase. The dispersed cells from rats treated with PGF-2α and were incubated in 10 mm glutamine+10 mm spermidine for 1 hour and for a further 5 hours in 10 mm glutamine without spermidine (lane 2) or in 10 mm glutamine+10 mm spermidine for 1 hour and for a further 5 hours in 10 mm glutamine+1 mm spermidine (lane 3). Control cells from rats treated with saline were dispersed with collagenase and incubated for 1 hour and a further 5 hours in glutamine only (lane 1). Five hundred nanograms of DNA from each sample was labeled with [α-32P]-dCTP using klenow enzyme, separated on a 1.8% agarose gel, and exposed to film for 24 hours. Results are shown in FIG. 18.
  • In yet another experiment, superovulated rats were injected subcutaneously with 1 mg/100 g body weight of spermidine, delivered in three equal doses of 0.333 mg/100 g body weight, 24, 12, and 2 hours prior to a subcutaneous injection with 500 μg PGF-2α. Control rats were divided into three sets: no injections, three injections of spermidine but no PGF-2α; and three injections with an equivalent volume of saline prior to PGF-2α treatment. Ovaries were removed front the rats either 1 hour and 35 minutes or 3 hours and 45 minutes after prostaglandin treatment and used for the isolation of DNA. Five hundred nanograms of DNA from each sample was labeled with [α-32P]-dCTP using Klenow enzyme, separated on a 1.8% agarose gel, and exposed to film for 24 hours: lane 1, no injections (animals were sacrificed at the same time as for lanes 3-5); lane 2, three injections with spermidine (animals were sacrificed at the same time as for lanes 3-5); lane 3, three injections with saline followed by injection with PGF-2α (animals were sacrificed 1 h and 35 min after treatment with PGF-2α); lane 4, three injections with spermidine followed by injection with PGF-2α (animals were sacrificed 1 h and 35 min after treatment with PGF-2α); lane 5, three injections with spermidine followed by injection with PGF-2α (animals were sacrificed 1 h and 35 min after treatment with PGF-2α); lane 6, three injections with spermidine followed by injection with PGF-2α (animals were sacrificed 3 h and 45 min after treatment with PGF-2α); lane 7, three injections with spermidine followed by injection with PGF-2α (animals were sacrificed 3 h and 45 min after treatment with PGF-2α). Results are shown in FIG. 19.
  • RNA Isolation
  • Total RNA was isolated from corpus luteum tissue removed from rats at various times after PGF-2α induction of apoptosis. Briefly, the tissue (5 g) was ground in liquid nitrogen. The ground powder was mixed with 30 ml guanidinium buffer (4 M guanidinium isothiocyanate, 2.5 mM NaOAc pH 8.5, 0.8% β-mercaptoethanol). The mixture was filtered through four layers of Miracloth and centrifuged at 10,000 g at 4° C. for 30 minutes. The supernatant was then subjected to cesium chloride density gradient centrifugation at 11,200 g for 20 hours. The pelleted RNA was rinsed with 75% ethanol, resuspended in 600 ml DEPC-treated water and the RNA precipitated at −70° C. with 1.5 ml 95% ethanol and 60 ml of 3M NaOAc.
  • Genomic DNA Isolation and Laddering
  • Genomic DNA was isolated from extracted corpus luteum tissue or dispersed corpus luteal cells using the QIAamp DNA Blood Kit (Qiagen) according to the manufacturer's instructions. The DNA was end-labeled by incubating 500 ng of DNA with 0.2 μCi[α-32P]dCTP, 1 mM Tris, 0.5 mM EDTA, 3 units of Klenow enzyme, and 0.2 pM each of dATP, dGTP, and dTTP, at room temperature for 30 minutes. Unincorporated nucleotides were removed by passing the sample through a 1-ml Sephadex G-50 column according to the method described by Maniatis et al. The samples were then resolved by Tris-acetate-EDTA (2%) gel electrophoresis. The gel was dried for 30 minutes at room temperature under vacuum and exposed to x-ray film at −80° C. for 24 hours.
  • Plasmid DNA Isolation, DNA Sequencing
  • The alkaline lysis method described by Sambrook et al., supra, was used to isolate plasmid DNA. The full-length positive cDNA clone was sequenced using the dideoxy sequencing method. Sanger et al., Proc. Natl. Acad. Sci. USA, 74:5463-5467. The open reading frame was compiled and analyzed using BLAST search (GenBank, Bethesda, Md.) and sequence alignment was achieved using a BCM Search Launcher: Multiple Sequence Alignments Pattern-Induced Multiple Alignment Method (see F. Corpet, Nuc. Acids Res., 16:10881-10890, (1987). Sequences and sequence alignments are shown in FIGS. 5-11.
  • Northern Blot Hybridization ofRat Corpus Luteum RNA
  • Twenty milligrams of total RNA isolated from rat corpus luteum at various stages of apoptosis were separated on 1% denatured formaldehyde agarose gels and immobilized on nylon membranes. The full-length rat apoptosis-specific eIF-5A cDNA (SEQ ID NO:1) labeled with 32P-dCTP using a random primer kit (Boehringer) was used to probe the membranes 7×107. Alternatively, full length rat DHS cDNA (SEQ ID NO:6) labeled with 32P-dCTP using a random primer kit (Boehringer) was used to probe the membranes (7×107 cpm). The membranes were washed once with 1×SSC, 0.1% SDS at room temperature and three times with 0.2×SSC, 0.1% SDS at 65° C. The membranes were dried and exposed to X-ray film overnight at −70° C.
  • As can be seen, apoptosis-specific eIF-5A and DHS are both upregulated in apoptosing corpus luteum tissue. Expression of apoptosis-specific eIF-5A is significantly enhanced after induction of apoptosis by treatment with PGF-2α—low at time zero, increased substantially within 1 hour of treatment, increased still more within 8 hours of treatment and increased slightly within 24 hours of treatment (FIG. 14). Expression of DHS was low at time zero, increased substantially within 1 hour of treatment, increased still more within 8 hours of treatment and increased again slightly within 24 hours of treatment (FIG. 15).
  • Generation of an Apoptosing Rat Corpus Luteum RT-PCR Product Using Primers Based on Yeast, Fungal and Human eIF-5A Sequences
  • A partial-length apoptosis-specific eIF-5A sequence (SEQ ID NO:11) corresponding to the 3′ end of the gene was generated from apoptosing rat corpus luteum RNA template by RT-PCR using a pair of oligonucleotide primers designed from yeast, fungal and human apoptosis-specific eIF-5A sequences. The upstream primer used to isolate the 3′end of the rat apoptosis-specific eIF-5A gene is a 20 nucleotide degenerate primer: 5′ TCSAARACHGGNAAGCAYGG 3′ (SEQ ID NO:9), wherein S is selected from C and G; R is selected from A and G; H is selected from A, T, and C; Y is selected from C and T; and N is any nucleic acid. The downstream primer used to isolate the 3′end of the rat eIF-5A gene contains 42 nucleotides: 5′ GCGAAGCTTCCATGG CTCGAGTTTTTTTTTTTTTTTTTTTTT 3′ (SEQ ID NO: 10). A reverse transcriptase polymerase chain reaction (RT-PCR) was carried out. Briefly, using 5 mg of the downstream primer, a first strand of cDNA was synthesized. The first strand was then used as a template in a RT-PCR using both the upstream and downstream primers.
  • Separation of the RT-PCR products on an agarose gel revealed the presence a 900 bp fragment, which was subcloned into pBluescript™ (Stratagene Cloning Systems, LaJolla, Calif.) using blunt end ligation and sequenced (SEQ ID NO:11). The cDNA sequence of the 3′ end is SEQ ID NO:11 and the amino acid sequence of the 3′ end is SEQ ID NO:12. See FIGS. 1-2.
  • A partial-length apoptosis-specific eIF-5A sequence (SEQ ID NO:15) corresponding to the 5′ end of the gene and overlapping with the 3′ end was generated from apoptosing rat corpus luteum RNA template by RT-PCR. The 5′ primer is a 24-mer having the sequence, 5′ CAGGTCTAGAGTTGGAATCGAAGC 3′ (SEQ ID NO:13), that was designed from human eIF-5A sequences. The 3′ primer is a 30-mer having the sequence, 5′ ATATCTCGAGCCTT GATTGCAACAGCTGCC 3′ (SEQ ID NO:14) that was designed according to the 3′ end RT-PCR fragment. A reverse transcriptase-polymerase chain reaction (RT-PCR) was carried out. Briefly, using 5 mg of the downstream primer, a first strand of cDNA was synthesized. The first strand was then used as a template in a RT-PCR using both the upstream and downstream primers.
  • Separation of the RT-PCR products on an agarose gel revealed the presence a 500 bp fragment, which was subcloned into pBluescript™ (Stratagene Cloning Systems, LaJolla, Calif.) using XbaI and XhoI cloning sites present in the upstream and downstream primers, respectively, and sequenced (SEQ ID NO:15). The cDNA sequence of the 5′ end is SEQ ID NO:15, and the amino acid sequence of the 5′ end is SEQ ID NO:16. See FIG. 2.
  • The sequences of the 3′ and 5′ ends of the rat apoptosis-specific eIF-5A (SEQ ID NO:11 and SEQ ID NO:15, respectively) overlapped and gave rise to the full-length cDNA sequence (SEQ ID NO:1). This full-length sequence was aligned and compared with sequences in the GeneBank data base. See FIGS. 1-2. The cDNA clone encodes a 154 amino acid polypeptide (SEQ ID NO:2) having a calculated molecular mass of 16.8 KDa. The nucleotide sequence, SEQ ID NO:1, for the full length cDNA of the rat apoptosis-specific corpus luteum eIF-5A gene obtained by RT-PCR is depicted in FIG. 3 and the corresponding derived amino acid sequence is SEQ ID NO:9. The derived full-length amino acid sequence of eIF-5A was aligned with human and mouse eIF-5a sequences. See FIG. 7-9.
  • Generation of an Apoptosing Rat Corpus Luteum RT-PCR Product Using Primers Based on a Human DHS Sequence
  • A partial-length DHS sequence (SEQ ID NO:6) corresponding to the 3′ end of the gene was generated from apoptosing rat corpus luteum RNA template by RT-PCR using a pair of oligonucleotide primers designed from a human DHS sequence. The 5′ primer is a 20-mer having the sequence, 5′ GTCTGTGTATTATTGGGCCC 3′ (SEQ ID NO. 17); the 3′ primer is a 42-mer having the sequence, 5′ GCGAAGCTTCCATGGC TCGAGTTTTTTTTTTTTTTTTTTTTT 3′ (SEQ ID NO:18). A reverse transcriptase polymerase chain reaction (RT-PCR) was carried out. Briefly, using 5 mg of the downstream primer, a first strand of cDNA was synthesized. The first strand was then used as a template in a RT-PCR using both the upstream and downstream primers.
  • Separation of the RT-PCR products on an agarose gel revealed the presence a 606 bp fragment, which was subcloned into pBluescript™ (Stratagene Cloning Systems, LaJolla, Calif.) using blunt end ligation and sequenced (SEQ ID NO:6). The nucleotide sequence (SEQ ID NO:6) for the partial length cDNA of the rat apoptosis-specific corpus luteum DHS gene obtained by RT-PCR is depicted in FIG. 4 and the corresponding derived amino acid sequence is SEQ ID NO.7.
  • Isolation of Genomic DNA and Southern Analysis
  • Genomic DNA for southern blotting was isolated from excised rat ovaries. Approximately 100 mg of ovary tissue was divided into small pieces and placed into a 15 ml tube. The tissue was washed twice with 1 ml of PBS by gently shaking the tissue suspension and then removing the PBS using a pipette. The tissue was resuspended in 2.06 ml of DNA-buffer (0.2 M Tris-HCl pH 8.0 and 0.1 mM EDTA) and 240 μl of 10% SDS and 100 μl of proteinase K (Boehringer Manheim; 10 mg/ml) was added. The tissue was placed in a shaking water bath at 45° C. overnight. The following day another 100 μl of proteinase K (10 mg/ml) was added and the tissue suspension was incubated in a water-bath at 45° C. for an additional 4 hours. After the incubation the tissue suspension was extracted once with an equal volume of phenol:chloroform:iso-amyl alcohol (25:24:1) and once with an equal volume of chloroform:iso-amyl alcohol (24:1). Following the extractions 1/10th volume of 3M sodium acetate (pH 5.2) and 2 volumes of ethanol were added. A glass pipette sealed and formed into a hook using a Bunsen burner was used to pull the DNA threads out of solution and to transfer the DNA into a clean microcentrifuge tube. The DNA was washed once in 70% ethanol and air-dried for 10 minutes. The DNA pellet was dissolved in 500 μl of 10 mM Tris-HCl (pH 8.0), 10 μl of RNase A (10 mg/ml) was added, and the DNA was incubated for 1 hour at 37° C. The DNA was extracted once with phenol:chloroforrn:iso-amyl alcohol (25:24:1) and the DNA was precipitated by adding 1/10th volume of 3 M sodium acetate (pH 5.2) and 2 volumes of ethanol. The DNA was pelleted by centrifugation for 10 minutes at 13,000×g at 4° C. The DNA pellet was washed once in 70% ethanol and dissolved in 200 μl 10 mM Tris-HCl (pH 8.0) by rotating the DNA at 4° C. overnight.
  • For Southern blot analysis, genomic DNA isolated from rat ovaries was digested with various restriction enzymes that either do not cut in the endogenous gene or cut only once. To achieve this, 10 μg genomic DNA, 20 μl 10× reaction buffer and 100 U restriction enzyme were reacted for five to six hours in a total reaction volume of 200 μl. Digested DNA was loaded onto a 0.7% agarose gel and subjected to electrophoresis for 6 hours at 40 volts or overnight at 15 volts. After electrophoresis, the gel was depurinated for 10 minutes in 0.2 N HCl followed by two 15-minute washes in denaturing solution (0.5 M NaOH, 1.5 M NaCl) and two 15 minute washes in neutralizing buffer (1.5 M NaCl, 0.5 M Tris-HCl pH 7.4). The DNA was transferred to a nylon membrane, and the membrane was prehybridized in hybridization solution (40% formamide, 6×SSC, 5× Denhart's, solution (1× Denhart's solution is 0.02% Ficoll, 0.02% PVP, and 0.02% BSA), 0.5% SDS, and 1.5 mg of denatured salmon sperm DNA). A 700 bp PCR fragment of the 3′ UTR of rat eIF-5A cDNA (650 bp of 3′ UTR and 50 bp of coding) was labeled with [a-32P]-dCTP by random priming and added to the membrane at 1×106 cpm/ml.
  • Similarly, a 606 bp PCR fragment of the rat DHS cDNA (450 bp coding and 156 bp 3′ UTR) was random prime labeled with [α-32P]-dCTP and added at 1×10 6 cpm/ml to a second identical membrane. The blots were hybridized overnight at 42° C. and then washed twice with 2×SSC and 0.1% SDS at 42° C. and twice with 1×SSC and 0.1% SDS at 42° C. The blots were then exposed to film for 3-10 days.
  • Rat corpus genomic DNA was cut with restriction enzymes as indicated on FIG. 20 and probed with 32P-dCTP-labeled full-length eIF-5A cDNA. Hybridization under high stringency conditions revealed hybridization of the full-length cDNA probe to several restriction fragments for each restriction enzyme digested DNA sample, indicating the presence of several isoforms of eIF-5A. Of particular note, when rat genomic DNA was digested with EcoRV, which has a restriction site within the open reading frame of apoptosis-specific eIF-5A, two restriction fragments of the apoptosis-specific isoform of eIF-5A were detectable in the Southern blot. The two fragments are indicated with double arrows in FIG. 20. The restriction fragment corresponding to the apoptosis-specific isoform of eIF-5A is indicated by a single arrow in the lanes labeled EcoRI and BamH1, restriction enzymes for which there are no cut sites within the open reading frame. These results suggest that the apoptosis-specific eIF-5A is a single copy gene in rat. As shown in FIGS. 5 through 13, the eIF-5A gene is highly conserved across species, and so it would be expected that there is a significant amount of conservation between isoforms within any species.
  • FIG. 21 shows a Southern blot of rat genomic DNA probed with 32P-dCTP-labeled partial-length rat corpus luteum DHS cDNA. The genomic DNA was cut with EcoRV, a restriction enzyme that does not cut the partial-length cDNA used as a probe. Two restriction fragments are evident indicating that there are two copies of the gene or that the gene contains an intron with an EcoRV site.
  • Example 2
  • The present example demonstrates modulation of apoptosis apoptosis-specific eIF-5A (increasing apoptosis with apoptosis-specific eIF-5A in sense orientation)
  • Culturing of COS-7 Cells and Isolation of RNA
  • COS-7, an African green monkey kidney fibroblast-like cell line transformed with a mutant of SV40 that codes for wild-type T antigen, was used for all transfection-based experiments. COS-7 cells were cultured in Dulbecco's Modified Eagle's medium (DMEM) with 0.584 grams per liter of L-glutamine, 4.5 g of glucose per liter, and 0.37% sodium bicarbonate. The culture media was supplemented with 10% fetal bovine serum (FBS) and 100 units of penicillin/streptomycin. The cells were grown at 37° C. in a humidified environment of 5% CO2 and 95% air. The cells were subcultured every 3 to 4 days by detaching the adherent cells with a solution of 0.25% trypsin and 1 mM EDTA. The detached cells were dispensed at a split ratio of 1:10 in a new culture dish with fresh media.
  • COS-7 cells to be used for isolation of RNA were grown in 150-mm tissue culture treated dishes (Coming). The cells were harvested by detaching them with a solution of trypsin-EDTA. The detached cells were collected in a centrifuge tube, and the cells were pelleted by centrifugation at 3000 rpm for 5 minutes. The supernatant was removed, and the cell pellet was flash-frozen in liquid nitrogen. RNA was isolated from the frozen cells using the GenElute Mammalian Total RNA Miniprep kit (Sigma) according to the manufacturer's instructions.
  • Construction of Recombinant Plasmids and Transfection of COS-7 Cells
  • Recombinant plasmids carrying the full-length coding sequence of rat apoptosis-specific eIF-5A in the sense orientation and the 3′ untranslated region (UTR) of rat apoptosis-specific eIF-5A in the antisense orientation were constructed using the mammalian epitope tag expression vector, pHM6 (Roche Molecular Biochemicals), which is illustrated in FIG. 21. The vector contains the following: CMV promoter—human cytomegalovirus immediate-early promoter/enhancer; HA—nonapeptide epitope tag from influenza hemagglutinin; BGH pA—Bovine growth hormone polyadenylation signal; f1 ori—f1 origin; SV40 ori—SV40 early promoter and origin; Neomycin—Neomycin resistance (G418) gene; SV40 pA—SV40 polyadenylation signal; Col E1—ColE1 origin; Ampicillin—Ampicillin resistance gene. The full-length coding sequence of rat apoptosis-specific eIF-5A and the 3′ UTR of rat apoptosis-specific eIF-5A were amplified by PCR from the original rat eIF-5A RT-PCR fragment in pBluescript (SEQ ID NO:1). To amplify the full-length eIF-5A the primers used were as follows: Forward 5′ GCCAAGCTTAATGGCAGATGATTT GG 3′ (SEQ ID NO: 59) (Hind3) and Reverse 5′ CTGAATTCCAGT TATTTTGCCATGG 3′ (SEQ ID NO:60) (EcoR1). To amplify the 3′ UTR rat apoptosis-specific eIF-5A the primers used were as follows: forward 5′ AATGAATTCCGCCATGACAGAGGAGGC 3′ (SEQ ID NO: 61) (EcoR1) and reverse 5′ GCGAAGCTTCCATGGCTCGAGTTTTTTTTTTTTTTTTTTTTT 3′ (SEQ ID NO: 62) (Hind3).
  • The full-length rat apoptosis-specific eIF-5A PCR product isolated after agarose gel electrophoresis was 430 bp in length while the 3′ UTR rat apoptosis-specific eIF-5A PCR product was 697 bp in length. Both PCR products were subcloned into the Hind 3 and EcoR1 sites of pHM6 to create pHM6-full-length apoptosis-specific eIF-5A and pHM6-antisense 3′UTReIF-5A. The full-length rat apoptosis-specific eIF-5A PCR product was subcloned in frame with the nonapeptide epitope tag from influenza hemagglutinin (HA) present upstream of the multiple cloning site to allow for detection of the recombinant protein using an anti-[HA]-peroxidase antibody. Expression is driven by the human cytomegalovirus immediate-early promoter/enhancer to ensure high level expression in mammalian cell lines. The plasmid also features a neomycin-resistance (G418) gene, which allows for selection of stable transfectants, and a SV40 early promoter and origin, which allows episomal replication in cells expressing SV40 large T antigen, such as COS-7.
  • COS-7 cells to be used in transfection experiments were cultured in either 24 well cell culture plates (Corning) for cells to be used for protein extraction, or 4 chamber culture slides (Falcon) for cells to be used for staining. The cells were grown in DMEM media supplemented with 10% FBS, but lacking penicillin/streptomycin, to 50 to 70% confluency. Transfection medium sufficient for one well of a 24-well plate or culture slide was prepared by diluting 0.32 μg of plasmid DNA in 42.5 μl of serum-free DMEM and incubating the mixture at room temperature for 15 minutes. 1.6 μl of the transfection reagent, LipofectAMINE (Gibco, BRL), was diluted in 42.5 μl of serum-free DMEM and incubated for 5 minutes at room temperature. After 5 minutes the LipofectAMINE mixture was added to the DNA mixture and incubated together at room temperature for 30 to 60 minutes. The cells to be transfected were washed once with serum-free DMEM before overlaying the transfection medium and the cells were placed back in the growth chamber for 4 hours.
  • After the incubation, 0.17 ml of DMEM +20% FBS was added to the cells. The cells were the cultured for a further 40 hours before either being induced to undergo apoptosis prior to staining or harvested for Western blot analysis. As a control, mock transfections were also performed in which the plasmid DNA was omitted from the transfection medium.
  • Protein Extraction and Western Blotting
  • Protein was isolated for Western blotting from transfected cells by washing the cells twice in PBS (8 g/L NaCl, 0.2 g/L KCl, 1.44 g/L Na2HPO4, and 0.24 g/L KH2PO4) and then adding 150 μl of hot SDS gel-loading buffer (50 mM Tris-HCl pH 6.8, 100 mM dithiothreitol, 2% SDS, 0.1% bromophenol blue, and 10% glycerol). The cell lysate was collected in a microcentrifuge tube, heated at 95° C. for 10 minutes, and then centrifuged at 13,000×g for 10 minutes. The supernatant was transferred to a fresh microcentrifuge tube and stored at −20° C. until ready for use.
  • For Western blotting, 2.5 or 5 μg of total protein was separated on a 12% SDS-polyacrylamide gel. The separated proteins were transferred to a polyvinylidene difluoride membrane. The membrane was then incubated for one hour in blocking solution (5% skim milk powder, 0.02% sodium azide in PBS) and washed three times for 15 minutes in PBS-T (PBS+0.05% Tween-20). The membrane was stored overnight in PBS-T at 4° C. After being warmed to room temperature the next day, the membrane was blocked for 30 seconds in 1 μg/ml polyvinyl alcohol. The membrane was rinsed 5 times in deionized water and then blocked for 30 minutes in a solution of 5% milk in PBS. The primary antibody was preincubated for 30 minutes in a solution of 5% milk in PBS prior to incubation with the membrane.
  • Several primary antibodies were used. An anti-[HA]-peroxidase antibody (Roche Molecular Biochemicals) was used at a dilution of 1:5000 to detect expression of the recombinant proteins. Since this antibody is conjugated to peroxidase, no secondary antibody was necessary, and the blot was washed and developed by chemiluminescence. The other primary antibodies that were used are monoclonal antibodies from Oncogene that recognize p53 (Ab-6), Bcl-2 (Ab-1), and c-Myc (Ab-2). The monoclonal antibody to p53 was used at a dilution of 0.1 μg/ml, and the monoclonal antibodies to Bcl-2 and c-Myc were both used at a dilution of 0.83 μg/ml. After incubation with primary antibody for 60 to 90 minutes, the membrane was washed 3 times for 15 minutes in PBS-T. Secondary antibody was then diluted in 1% milk in PBS and incubated with the membrane for 60 to 90 minutes. When p53 (Ab-6) was used as the primary antibody, the secondary antibody used was a goat anti-mouse IgG conjugated to alkaline phosphatase (Rockland) at a dilution of 1:1000. When Bcl-2 (Ab-1) and c-Myc (Ab-2) were used as the primary antibody, a rabbit anti-mouse IgG conjugated to peroxidase (Sigma) was used at a dilution of 1:5000. After incubation with the secondary antibody, the membrane was washed 3 times in PBS-T.
  • Two detection methods were used to develop the blots, a colorimetric method and a chemiluminescent method. The colorimetric method was used only when p53 (Ab-6) was used as the primary antibody in conjunction with the alkaline phosphatase-conjugated secondary antibody. Bound antibody was visualized by incubating the blot in the dark in a solution of 0.33 mg/mL nitro blue tetrazolium, 0.165 mg/mL 5-bromo-4-chloro-3-indolyl phosphate, 100 mM NaCl, 5 mM MgCl2, and 100 mM Tris-HCl (pH 9.5). The color reaction was stopped by incubating the blot in 2 mM EDTA in PBS. A chemiluminescent detection method was used for all other primary antibodies, including anti-[HA]-peroxidase, Bcl-2 (Ab-1), and c-Myc (Ab-2). The ECL Plus Western blotting detection kit (Amersham Pharmacia Biotech) was used to detect peroxidase-conjugated bound antibodies. In brief, the membrane was lightly blotted dry and then incubated in the dark with a 40:1 mix of reagent A and reagent B for 5 minutes. The membrane was blotted dry, placed between sheets of acetate, and exposed to X-ray film for time periods varying from 10 seconds to 10 minutes.
  • Induction ofApoptosis in COS 7 Cells
  • Two methods were used to induce apoptosis in transfected COS-7 cells, serum deprivation and treatment with Actinomycin D, streptomyces sp (Calbiochem). For both treatments, the medium was removed 40 hours post-transfection. For serum starvation experiments, the media was replaced with serum- and antibiotic-free DMEM. Cells grown in antibiotic-free DMEM supplemented with 10% FBS were used as a control. For Actinomycin D induction of apoptosis, the media was replaced with antibiotic-free DMEM supplemented with 10% FBS and 1 μg/ml Actinomycin D dissolved in methanol. Control cells were grown in antibiotic-free DMEM supplemented with 10% FBS and an equivalent volume of methanol. For both methods, the percentage of apoptotic cells was determined 48 hours later by staining with either Hoescht or Annexin V-Cy3. Induction of apoptosis was also confirmed by Northern blot analyses, as shown in FIG. 22.
  • Hoescht Staining
  • The nuclear stain, Hoescht, was used to label the nuclei of transfected COS-7 cells in order to identify apoptotic cells based on morphological features such as nuclear fragmentation and condensation. A fixative, consisting of a 3:1 mixture of absolute methanol and glacial acetic acid, was prepared immediately before use. An equal volume of fixative was added to the media of COS-7 cells growing on a culture slide and incubated for 2 minutes. The media/fixative mixture was removed from the cells and discarded, and 1 ml of fixative was added to the cells. After 5 minutes the fixative was discarded, and 1 ml of fresh fixative was added to the cells and incubated for 5 minutes. The fixative was discarded, and the cells were air-dried for 4 minutes before adding 1 ml of Hoescht stain (0.5 μg/ml Hoescht 33258 in PBS). After a 10-minute incubation in the dark, the staining solution was discarded and the slide was washed 3 times for 1 minute with deionized water. After washing, 1 ml of Mcllvaine's buffer (0.021 M citric acid, 0.058 M Na2HPO4.7H2O; pH 5.6) was added to the cells, and they were incubated in the dark for 20 minutes. The buffer was discarded, the cells were air-dried for 5 minutes in the dark and the chambers separating the wells of the culture slide were removed. A few drops of Vectashield mounting media for fluorescence (Vector Laboratories) was added to the slide and overlaid with a coverslip. The stained cells were viewed under a fluorescence microscope using a UV filter. Cells with brightly stained or fragmented nuclei were scored as apoptotic.
  • Annexin V-Cy3 Staining
  • An Annexin V-Cy3 apoptosis detection kit (Sigma) was used to fluorescently label externalized phosphatidylserine on apoptotic cells. The kit was used according to the manufacturer's protocol with the following modifications. In brief, transfected COS-7 cells growing on four chamber culture slides were washed twice with PBS and three times with 1× Binding Buffer. 150 μl of staining solution (1 μg/ml AnnCy3 in 1× Binding Buffer) was added, and the cells were incubated in the dark for 10 minutes. The staining solution was then removed, and the cells were washed 5 times with 1× Binding Buffer. The chamber walls were removed from the culture slide, and several drops of 1× Binding Buffer were placed on the cells and overlaid with a coverslip. The stained cells were analyzed by fluorescence microscopy using a green filter to visualize the red fluorescence of positively stained (apoptotic) cells. The total cell population was determined by counting the cell number under visible light.
  • Example 3
  • The present example demonstrates modulation of apoptosis apoptosis-specific eIF-5A.
  • Using the general procedures and methods described in the previous examples, FIG. 23 is a flow chart illustrating the procedure for transient transfection of COS-7 cells, in which cells in serum-free medium were incubated in plasmid DNA in lipofectAMINE for 4 hours, serum was added, and the cells were incubated for a further 40 hours. The cells were then either incubated in regular medium containing serum for a further 48 hours before analysis (i.e. no further treatment), deprived of serum for 48 hours to induce apoptosis before analysis, or treated with actinomycin D for 48 hours to induce apoptosis before analysis.
  • FIG. 22 is a Western blot illustrating transient expression of foreign proteins in COS-7 cells following transfection with pHM6. Protein was isolated from COS-7 cells 48 hours after either mock transfection, or transfection with pHM6-LacZ, pHM6-Antisense 3′ rF5A (pHM6-Antisense 3′ UTR rat apoptosis-specific eIF-5A), or pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A). Five μg of protein from each sample was fractionated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with anti-[HA]-peroxidase. The bound antibody was detected by chemiluminescence and exposed to x-ray film for 30 seconds. Expression of LacZ (lane 2) and of sense rat apoptosis-specific eIF-5A (lane 4) is clearly visible.
  • As described above, COS-7 cells were either mock transfected or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A). Forty hours after transfection, the cells were induced to undergo apoptosis by withdrawal of serum for 48 hours. The caspase proteolytic activity in the transfected cell extract was measured using a fluorometric homogenous caspase assay kit (Roche Diagnostics). DNA fragmentation was also measured using the FragEL DNA Fragmentation Apoptosis Detection kit (Oncogene) which labels the exposed 3′-OH ends of DNA fragments with fluorescein-labeled deoxynucleotides.
  • Additional COS-7 cells were either mock transfected or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A). Forty hours after transfection, the cells were either grown for an additional 48 hours in regular medium containing serum (no further treatment), induced to undergo apoptosis by withdrawal of serum for 48 hours or induced to undergo apoptosis by treatment with 0.5 μg/ml of Actinomycin D for 48 hours. The cells were either stained with Hoescht 33258, which depicts nuclear fragmentation accompanying apoptosis, or stained with Annexin V-Cy3, which depicts phosphatidylserine exposure accompanying apoptosis. Stained cells were also viewed by fluorescence microscopy using a green filter and counted to determine the percentage of cells undergoing apoptosis. The total cell population was counted under visible light.
  • FIG. 25 illustrates enhanced apoptosis as reflected by increased caspase activity when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specificeIF-5A in the sense orientation. Expression of rat apoptosis-specificeIF-5A resulted in a 60% increase in caspase activity.
  • FIG. 26 illustrates enhanced apoptosis as reflected by increased DNA fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specificeIF-5A in the sense orientation. Expression of rat apoptosis-specificeIF-5A resulted in a 273% increase in DNA fragmentation. FIG. 27 illustrates detection of apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation. There is a greater incidence of fragmented nuclei in cells expressing rat apoptosis-specificeIF-5A. FIG. 28 illustrates enhanced apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation. Expression of rat apoptosis-specificeIF-5A resulted in a 27% and 63% increase in nuclear fragmentation over control in non-serum starved and serum starved samples, respectively.
  • FIG. 29 illustrates detection of apoptosis as reflected by phosphatidylserine exposure when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation. FIG. 30 illustrates enhanced apoptosis as reflected by increased phosphatidylserine exposure when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation. Expression of rat apoptosis-specific eIF-5A resulted in a 140% and 198% increase in phosphatidylserine exposure over control, in non-serum starved and serum starved samples, respectively.
  • FIG. 31 illustrates enhanced apoptosis as reflected by increased nuclear fragmentation when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specificeIF-5A in the sense orientation. Expression of rat apoptosis-specific eIF-5A resulted in a 115% and 62% increase in nuclear fragmentation over control in untreated and treated samples, respectively. FIG. 32 illustrates a comparison of enhanced apoptosis under conditions in which COS-7 cells transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation were either given no further treatment or treatment to induce apoptosis.
  • Example 4
  • The present example demonstrates modulation of apoptotic activity following administration of apoptosis-specific eIF-5A.
  • COS-7 cells were either mock transfected, transfected with pHM6-LacZ or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A) and incubated for 40 hours. Five μg samples of protein extract from each sample were fractionated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with a monoclonal antibody that recognizes Bcl-2. Rabbit anti-mouse IgG conjugated to peroxidase was used as a secondary antibody, and bound antibody was detected by chemiluminescence and exposure to x-ray film. Results are shown in FIG. 33. Less Bcl-2 is detectable in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ; thus showing that Bcl-2 is down-regulated with the pHM6-sense rF5A construct.
  • Additional COS-7 cells were either mock transfected, transfected with pHM6-antisense 3′ rF5A (pHM6-antisense 3′ UTR of rat apoptosis-specific eIF-5A) or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A). Forty hours after transfection, the cells were induced to undergo apoptosis by withdrawal of serum for 48 hours. Five μg samples of protein extract from each sample were fractionated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with a monoclonal antibody that recognizes Bcl-2. Rabbit anti-mouse IgG conjugated to peroxidase was used as a secondary antibody, and bound antibody was detected by chemiluminescence and exposure to x-ray film.
  • Also additionally, COS-7 cells were either mock transfected, transfected with pHM6-LacZ or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A) and incubated for 40 hours. Five μg samples of protein extract from each sample were fractionated by SDS-PAGE, transferred to a PVDF membrane, and Western blotted with a monoclonal antibody that recognizes p53. Goat anti-mouse IgG conjugated to alkaline phosphatase was used as a secondary antibody, and bound antibody was detected a colorimetrically.
  • Finally, COS-7 cells were either mock transfected, transfected with pHM6-LacZ or transfected with pHM6-Sense rF5A (pHM6-Full length rat apoptosis-specific eIF-5A) and incubated for 40 hours. Five μg samples of protein extract from each sample were fractionated by SDS-PAGE, transferred to a PVDF membrane, and probed with a monoclonal antibody that recognizes p53. Corresponding protein blots were probed with anti-[HA]-peroxidase to determine the level of rat apoptosis-specific eIF-5A expression. Goat anti-mouse IgG conjugated to alkaline phosphatase was used as a secondary antibody, and bound antibody was detected by chemiluminescence.
  • FIG. 33 illustrates downregulation of Bcl-2 when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation. The upper panel illustrates the Coomassie-blue-stained protein blot; the lower panel illustrates the corresponding Western blot. Less Bcl-2 is detectable in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ.
  • FIG. 34 illustrates upregulation of Bcl-2 when COS-7 cells were transiently transfected with pHM6 containing the 3′ end of apoptosis-specific eIF-5A in the antisense orientation. The upper panel illustrates the Coomassie-blue-stained protein blot; the lower panel illustrates the corresponding Western blot. More Bcl-2 is detectable in cells transfected with pHM6-antisense 3′ rF5A than in those mock transfected or transfected with pHM6-Sense rF5A.
  • FIG. 35 illustrates upregulation of c-Myc when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation. The upper panel illustrates the Coomassie-blue-stained protein blot; the lower panel illustrates the corresponding Western blot. Higher levels of c-Myc is detected in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ or the mock control.
  • FIG. 36 illustrates upregulation of p53 when COS-7 cells were transiently transfected with pHM6 containing full-length rat apoptosis-specific eIF-5A in the sense orientation. The upper panel illustrates the Coomassie-blue-stained protein blot; the lower panel illustrates the corresponding Western blot. Higher levels of p53 is detected in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ or the mock control.
  • FIG. 37 illustrates the dependence of p53 upregulation upon the expression of pHM6-full length rat apoptosis-specificeIF-5A in COS-7 cells. More rat apoptosis-specificeIF-5A is detectable in the first transfection than in the second transfection. In the Western blot probed with anti-p53, the panel illustrates a corresponding Coomassie-blue-stained protein blot and the panel illustrates the Western blot with p53. For the first transfection, more p53 is detectable in cells transfected with pHM6-Sense rF5A than in those transfected with pHM6-LacZ or the mock control. For the second transfection in which there was less expression of rat apoptosis-specific eIF-5A, there was no detectable difference in levels of p53 between cells transfected with pHM6-Sense rF5A, pHM6-LacZ or the mock control.
  • Example 5
  • FIG. 47 depicts an experiment run on heart tissue to mimic the beating of a human heart and the subsequent induced heart attack. FIG. 49 shows the laboratory bench set up. A slice of human heart tissue removed during valve replacement surgery was hooked up to electrodes. A small weight was attached to the heart tissue to ease in measuring the strength of the heart beats. The electrodes provided an electrical stimulus to get the tissue to start beating. The levels of gene expression for both apoptosis-specific eIF-5A and proliferating eIF-5A were measured in the heart tissue before ischemia was induced. See FIG. 46. In the pre-ischemic heart tissue low levels both apoptosis-specific eIF-5A and proliferating eIF-5A were produced and their levels were in relative balance. During this time, oxygen and carbon dioxide were delivered in a buffer to the heart at 92.5% and 7.5%, respectively. Later, the oxygen levels was reduced and the nitrogen levels was increased, to induce ischemia and finally a “heart attack.” The heart tissue stopped beating. The oxygen levels were then returned to normal, the heart tissue was pulsed again with an electrical stimulus to start the heart beating again. After the “heart attack” the expression levels of apoptosis-specific eIF-5A and proliferating eIF-5A were again measured. This time, there was a significant increase in the level of expression of the apoptosis-specific eIF-5A levels, whereas the increase in the level of expression of proliferating eIF-5A was noticeably less. See FIG. 46.
  • After the “heart attack” the heart did not beat as strong, as indicated by less compression/movement of the attached weight, thus indicating that the heart tissue cells were being killed rapidly due. to the presence of apoptosis-specific eIF-5A.
  • The EKG results are depicted in FIG. 48. On the left side of the panels a normal heart beat is shown (the pre-ischemic heart tissue). After the “heart attack” (straight line), and the re-initiation of the heart beat, the EKG shows decreased activity due to muscle cell death. The EKG shows relative loss in strength of heart beat.
  • Example 6
  • The following examples provide cell culture conditions.
  • Human Lamina Cribrosa and Astrocyte Culture
  • Paired human eyes were obtained within 48 hours post mortem from the Eye Bank of Canada, Ontario Division. Optic nerve heads (with attached pole) were removed and placed in Dulbecco's modified Eagle's medium (DMEM) supplemented with antibiotic/antimycotic, glutamine, and 10% FBS for 3 hours. The optic nerve head (ONH) button was retrieved from each tissue sample and minced with fine dissecting scissors into four small pieces. Explants were cultured in 12.5 cm2 plastic culture flasks in DMEM medium. Growth was observed within one month in viable explants. Once the cells reached 90% confluence, they were trypsinized and subjected to differential subculturing to produce lamina cribrosa (LC) and astrocyte cell populations. Specifically, LC cells were subcultured in 25 cm2 flasks in DMEM supplemented with gentamycin, glutamine, and 10% FBS, whereas astrocytes were expanded in 25 cm2 flasks containing EBM complete medium (Clonetics) with no FBS. FBS was added to astrocyte cultures following 10 days of subculture. Cells were maintained and subcultured as per this protocol.
  • Cell populations obtained by differential subculturing were characterized for identity and population purity using differential fluorescent antibody staining on 8 well culture slides. Cells were fixed in 10% formalin solution and washed three times with Dulbecco's Phosphate Buffered Saline (DPBS). Following blocking with 2% nonfat milk in DPBS, antibodies were diluted in 1% BSA in DPBS and applied to the cells in 6 of the wells. The remaining two wells were treated with only 1% bovine serum albumin (BSA) solution and no primary antibody as controls. Cells were incubated with the primary antibodies for one hour at room temperature and then washed three times with DPBS. Appropriate secondary antibodies were diluted in 1% BSA in DPBS, added to each well and incubated for 1 hour. Following washing with DPBS, the chambers separating the wells of the culture slide were removed from the slide, and the slide was immersed in double distilled water and then allowed to air-dry. Fluoromount (Vector Laboratories) was applied to each slide and overlayed by 22×60 mm coverglass slips.
  • Immunofluorescent staining was viewed under a fluorescent microscope with appropriate filters and compared to the control wells that were not treated with primary antibody. All primary antibodies were obtained from Sigma unless otherwise stated. All secondary antibodies were purchased from Molecular Probes. Primary antibodies used to identify LC cells were: anti-collagen I, anti-collagen IV, anti-laminin, anti-cellular fibronectin. Primary antibodies used to identify astrocytes were: anti-galactocerebroside (Chemicon International), anti-A2B5 (Chemicon International), anti-NCAM, anti-human Von willebrand Factor. Additional antibodies used for both cell populations included anti-glial fibrillary (GFAP) and anti-alpha-smooth muscle actin. Cell populations were determined to be comprised of LC cells if they stained positively for collagen I, collagen IV, laminin, cellular fibronectin, alpha smooth muscle actin and negatively for glial fibrillary (GFAP). Cell populations were determined to be comprised of astrocytes if they stained positively for NCAM, glial fibrillary (GFAP), and negatively for galactocerebroside, A2B5, human Von willebrand Factor, and alpha smooth muscle actin.
  • In this preliminary study, three sets of human eyes were used to initiate cultures. LC cell lines #506, #517, and #524 were established from the optic nerve heads of and 83-year old male, a 17-year old male, and a 26-year old female, respectively. All LC cell lines have been fully characterized and found to contain greater than 90% LC cells.
  • RKO Cell Culture
  • RKO (American Type Culture Collection CRL-2577), a human colon carcinoma cell line expressing wild-type p53, was used to test the antisense oligonucleotides for the ability to suppress apoptosis-specific eIF-5A protein expression. RKO were cultured in Minimum Essential Medium Eagle (MEM) with non-essential amino acids, Earle's salts, and L-glutamine. The culture media was supplemented with 10% fetal bovine serum (FBS) and 100 units of penicillin/streptomycin. The cells were grown at 37° C. in a humidified environment of 5% CO2 and 95% air. The cells were subcultured every 3 to 4 days by detaching the adherent cells with a solution of 0.25% trypsin and 1 mM EDTA. The detached cells were dispensed at a split ratio of 1:10 to 1:12 into a new culture dish with fresh media.
  • HepG2 Cell Culture
  • HepG2, a human hepatocellular carcinoma cell line, was used to test the ability of an antisense oligo directed against human apoptosis-specific eIF-5A to block production of TNF-α in response to treatment with IL-1α. HepG2 cells were cultured in DMEM supplemented with gentamycin, glutamine, and 10% FBS and grown at 37° C. in a humidified environment of 5% CO2 and 95% air.
  • Example 7
  • Induction of Apoptosis
  • Apoptosis was induced in RKO and lamina cribrosa cells using Actinomycin D, an RNA polymerase inhibitor, and camptothecin, a topoisomerase inhibitor, respectively. Actinomycin D was used at a concentration of 0.25 μg/ml and camptothecin was used at a concentration of 20, 40, or 50 μM. Apoptosis was also induced in lamina cribrosa cells using a combination of camptothecin (50 μM) and TNF-α (10 ng/ml). The combination of camptothecin and TNF-α was found to be more effective at inducing apoptosis than either camptothecin or TNF-α alone.
  • Antisense Oligonucleotides
  • A set of three antisense oligonucleotides targeted against human apoptosis-specific eIF-5A were designed by, and purchased from, Molecula Research Labs. The sequence of the first antisense oligonucleotide targeted against human apoptosis-specific eIF-5A (#1) was 5′ CCT GTC TCG AAG TCC AAG TC 3′ (SEQ ID NO: 63). The sequence of the second antisense oligonucleotide targeted against human apoptosis-specific eIF-5A (#2) was 5′ GGA CCT TGG CGT GGC CGT GC 3′ (SEQ ID NO: 64). The sequence of the third antisense oligonucleotide targeted against human apoptosis-specific eIF-5A (#3) was 5′ CTC GTA CCT CCC CGC TCT CC 3′ (SEQ ID NO: 65). The control oligonucleotide had the sequence 5′ CGT ACC GGT ACG GTT CCA GG 3′ (SEQ ID NO: 66). A fluorescein isothiocyanate (FITC)-labeled antisense oligonucleotide (Molecula Research Labs) was used to monitor transfection efficiency and had the sequence 5′ GGA CCT TGG CGT GGC CGT GCX 3′ (SEQ ID NO: 67), where X is the FITC label. All antisense oligonucleotides were fully phosphorothioated.
  • Transfection of Antisense Oligonucleotides
  • The ability of the apoptosis-specific eIF-5A antisense oligonucleotides to block apoptosis-specific eIF-5A protein expression was tested in RKO cells. RKO cells were transfected with antisense oligonucleotides using the transfection reagent, Oligofectamine (Invitrogen). Twenty four hours prior to transfection, the cells were split onto a 24 well plate at 157,000 per well in MEM media supplemented with 10% FBS but lacking penicillin/streptomycin. Twenty four hours later the cells had generally reached a confluency of approximately 50%. RKO cells were either mock transfected, or transfected with 100 nM or 200 nM of antisense oligonucleotide. Transfection medium sufficient for one well of a 24 well plate was prepared by diluting 0, 1.25, or 2.5 μl of a 20 μM stock of antisense oligonucleotide with serum-free MEM to a final volume of 42.5 μl and incubating the mixture at room temperature for 15 minutes. 1.5 μl of Oligofectamine was diluted in 6 μl of serum-free MEM and incubated for 7.5 minutes at room temperature. After 5 minutes the diluted Oligofectamine mixture was added to the DNA mixture and incubated together at room temperature for 20 minutes. The cells were washed once with serum-free MEM before adding 200 μl of MEM to the cells and overlaying 50 μl of transfection medium. The cells were placed back in the growth chamber for 4 hours. After the incubation, 125 μl of MEM +30% FBS was added to the cells. The cells were then cultured for a further 48 hours, treated with 0.25 μg/ml Actinomycin D for 24 hours, and then cell extract was harvested for Western blot analysis.
  • Transfection of lamina cribrosa cells was also tested using 100 and 200 nM antisense oligonucleotide and Oligofectamine using the same procedure described for RKO cells. However, effective transfection of lamina cribrosa cells was achieved by simply adding antisense oligonucleotide, diluted from 1 μM to 10 μM in serum-free media, to the cells for 24 hours and thereafter replacing the media with fresh antisense oligonucleotides diluted in serum-containing media every 24 hours for a total of two to five days.
  • The efficiency of antisense oligonucleotide transfection was optimized and monitored by performing transfections with an FITC-labeled antisense oligonucleotide having the same sequence as apoptosis-specific eIF-5A antisense oligonucleotide #2 (SEQ ID NO:64) but conjugated to FITC at the 3′ end. RKO and lamina cribrosa cells were transfected with the FITC-labeled antisense oligonucleotide on an 8-well culture slide. Forty-eight hours later the cells were washed with PBS and fixed for 10 minutes in 3.7% formaldehyde in PBS. The wells were removed and mounting media (Vectashield) was added, followed by a coverslip. The cells were then visualized under UV light on a fluorescent microscope nucleus using a fluorescein filter (Green H546, filter set 48915) and cells fluorescing bright green were determined to have taken up the oligonucleotide.
  • Detection of Apoptosis
  • Following transfection of lamina cribosa cells with antisense oligonucleotides and induction of apoptosis with camptothecin, the percentage of cells undergoing apoptosis in cells treated with either control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A SEQ ID NO:26 was determined. Two methods were used to detect apoptotic lamina cribosa cells—Hoescht staining and DeadEnd™ Fluorometric TUNEL. The nuclear stain, Hoescht, was used to label the nuclei of lamina cribosa cells in order to identify apoptotic cells based on morphological features such as nuclear fragmentation and condensation. A fixative, consisting of a 3:1 mixture of absolute methanol and glacial acetic acid, was prepared immediately before use. An equal volume of fixative was added to the media of cells growing on a culture slide and incubated for 2 minutes. The media/fixative mixture was removed from the cells and discarded and 1 ml of fixative was added to the cells. After 5 minutes the fixative was discarded and 1 ml of fresh fixative was added to the cells and incubated for 5 minutes. The fixative was discarded and the cells were air-dried for 4 minutes before adding 1 ml of Hoescht stain (0.5 μg/ml Hoescht 33258 in PBS). After a 10 minute incubation in the dark, the staining solution was discarded, the chambers separating the wells of the culture slide were removed, and the slide was washed 3 times for 1 minute with deionized water. After washing, a few drops of Mcllvaine's buffer (0.021 M citric acid, 0.058 M Na2HPO4.7H2O; pH 5.6) was added to the cells and overlaid with a coverslip. The stained cells were viewed under a fluorescent microscope using a UV filter. Cells with brightly stained or fragmented nuclei were scored as apoptotic. A minimum of 200 cells were counted per well.
  • The DeadEnd™ Fluorometric TUNEL (Promega) was used to detect the DNA fragmentation that is a characteristic feature of apoptotic cells. Following Hoescht staining, the culture slide was washed briefly with distilled water, and further washed by immersing the slide twice for 5 minutes in PBS (137 mM NaCl, 2.68 mM KCl, 1.47 mM KH2PO4, 8.1 mM Na2HPO4), blotting the slide on paper towel between washes. The cells were permeabilized by immersing them in 0.2% Triton X-100 in PBS for 5 minutes. The cells were then washed again by immersing the slide twice for 5 minutes in PBS and blotting the slide on paper towel between washes. 25 μl of equilibration buffer [200 mM potassium cacodylate (pH 6.6), 25 mM Tris-HCl (pH 6.6), 0.2 mM dithiothreitol, 0.25 mg/ml bovine serum albumin, and 2.5 mM cobalt chloride] was added per well and incubated for 5 to 10 minutes. During equilibration, 30 μl of reaction mixture was prepared for each well by mixing in a ratio of 45:5:1, respectively, equilibration buffer, nucleotide mix [50 μM fluorescein-12-dUTP, 100 μM dATP, 10 mM Tris-HCl (pH 7.6), and 1 mM EDTA], and terminal deoxynucleotidyl transferase enzyme (Tdt, 25 U/μl). After the incubation in equilibration buffer, 30 μl of reaction mixture was added per well and overlayed with a coverslip. The reaction was allowed to proceed in the dark at 37° C. for 1 hour. The reaction was terminated by immersing the slide in 2×SSC [0.3 M NaCl, and 30 mM sodium citrate (pH 7.0)] and incubating for 15 minutes. The slide was then washed by immersion in PBS three times for 5 minutes. The PBS was removed by sponging around the wells with a Kim wipe, a drop of mounting media (Oncogene research project, JA1750-4ML) was added to each well, and the slide was overlayed with a coverslip. The cells were viewed under a fluorescent microscope using a UV filter (UV-G 365, filter set 487902) in order to count the Hoescht-stained nuclei. Any cells with brightly stained or fragmented nuclei were scored as apoptotic. Using the same field of view, the cells were then viewed using a fluorescein filter (Green H546, filter set 48915) and any nuclei fluorescing bright green were scored as apoptotic. The percentage of apoptotic cells in the field of view was calculated by dividing the number of bright green nuclei counted using the fluorescein filter by the total number of nuclei counted under the UV filter. A minimum of 200 cells were counted per well.
  • FIGS. 54-57 depict the results of these studies. The percentage of apoptotic cells in samples having been transfected with apoptosis-specific eIF-5A is clearly much less than seen in cells having been transfected with the control oligonucleotide.
  • Protein Extraction and Western Blotting
  • Protein from transfected RKO cells was harvested for Western blot analysis by washing the cells with PBS, adding 40 μl of hot lysis buffer [0.5% SDS, 1 mM dithiothreitol, 50 mM Tris-HCl (pH 8.0)] per well. The cells were scraped and the resulting extract was transferred to a microfuge tube, boiled for 5 minutes, and stored at—20° C. The protein was quantitated using the Bio-Rad Protein Assay (Bio-Rad) according to the manufacturer's instructions.
  • For Western blotting 5 μg of total protein was separated on a 12% SDS-polyacrylamide gel. The separated proteins were transferred to a potyvinylidene difluoride membrane. The membrane was then incubated for one hour in blocking solution (5% skim milk powder in PBS) and washed three times for 15 minutes in 0.05% Tween-20/PBS. The membrane was stored overnight in PBS-T at 4° C. After being warmed to room temperature the next day, the membrane was blocked for 30 seconds in 1 μg/ml polyvinyl alcohol. The membrane was rinsed 5 times in deionized water and then blocked for 30 minutes in a solution of 5% milk in 0.025% Tween-20/PBS. The primary antibody was preincubated for 30 minutes in a solution of 5% milk in 0.025% Tween-20/PBS prior to incubation with the membrane.
  • Several primary antibodies were used. A monoclonal antibody from Oncogene which recognizes p53 (Ab-6) and a polyclonal antibody directed against a synthetic peptide (amino-CRLPEGDLGKEIEQKYD-carboxy) (SEQ ID NO:68) homologous to the c-terminal end of human apoptosis-specific eIF-5A that was raised in chickens (Gallus Immunotech). An anti-β-actin antibody (Oncogene) was also used to demonstrate equal loading of protein. The monoclonal antibody to p53 was used at a dilution of 0.05 μg/ml, the antibody against apoptosis-specific eIF-5A was used at a dilution of 1:1000, and the antibody against actin was used at a dilution of 1:20,000. After incubation with primary antibody for 60 to 90 minutes, the membrane was washed 3 times for 15 minutes in 0.05% Tween-20/PBS. Secondary antibody was then diluted in 1% milk in 0.025% Tween-20/PBS and incubated with the membrane for 60 to 90 minutes. When p53 (Ab-6) was used as the primary antibody, the secondary antibody used was a rabbit anti-mouse IgG conjugated to peroxidase (Sigma) at a dilution of 1:5000. When anti-apoptosis-specific eIF-5A was used as the primary antibody, a rabbit anti-chicken IgY conjugated to peroxidase (Gallus Immunotech) was used at a dilution of 1:5000. The secondary antibody used with actin was a goat anti-mouse IgM conjugated to peroxidase (Calbiochem) used at a dilution of 1:5000. After incubation with the secondary antibody, the membrane was washed 3 times in PBS-T.
  • The ECL Plus Western blotting detection kit (Amersham Pharmacia Biotech) was used to detect peroxidase-conjugated bound antibodies. In brief, the membrane was lightly blotted dry and then incubated in the dark with a 40:1 mix of reagent A and reagent B for 5 minutes. The membrane was blotted dry, placed between sheets of acetate, and exposed to X-ray film for time periods varying from 10 seconds to 30 minutes. The membrane was stripped by submerging the membrane in stripping buffer [100 mM 2-Mercaptoethanol, 2% SDS, and 62.5 mM Tris-HCl (pH 6.7)], and incubating at 50° C. for 30 minutes. The membrane was then rinsed in deionized water and washed twice for 10 minutes in large volumes of 0.05% Tween-20/PBS. Membranes were stripped and re-blotted up to three times.
  • Example 8
  • Construction of siRNA
  • Small inhibitory RNAs (siRNAs) directed against human apoptosis-specific eIF-5A were used to specifically suppress expression of apoptosis-specific eIF-5A in RKO and lamina cribrosa cells. Six siRNAs were generated by in vitro transcription using the Silencer™ siRNA Construction Kit (Ambion Inc.). Four siRNAs were generated against human apoptosis-specific eIF-5A (siRNAs #1 to #4)(SEQ ID NO:30-33). Two siRNAs were used as controls; an siRNA directed against GAPDH provided in the kit, and an siRNA (siRNA #5)(SEQ ID NO: 34) which had the reverse sequence of the apoptosis-specific eIF-5A siRNA #1 (SEQ ID NO:30) but does not itself target apoptosis-specific eIF-5A. The siRNAs were generated according to the manufacturer's protocol. In brief, DNA oligonucleotides encoding the desired siRNA strands were used as templates for T7 RNA polymerase to generate individual strands of the siRNA following annealing of a T7 promoter primer and a fill-in reaction with Klenow fragment. Following transcription reactions for both the sense and antisense strands, the reactions were combined and the two siRNA strands were annealed, treated with DNase and RNase, and then column purified. The sequence of the DNA oligonucleotides (T7 primer annealing site underlined) used to generate the siRNAs were: siRNA #1 antisense 5′ AAAGGAATGACTTCCAGCTGACCTGTCTC 3′ (SEQ ID NO:69) and siRNA #1 sense 5′ AATCAGCTGGAAGTCATTCCTCCTGTCTC 3′ (SEQ ID NO:70); siRNA #2 antisense 5′ AAGATCGTCGAGATGTCTACTCCTGTCTC 3′ (SEQ ID NO:71) and siRNA #2 sense 5′ AAAGTAGACATCTCGACGATCCCTGTCTC 3′ (SEQ ID NO:72); siRNA #3 antisense 5′ AAGGTCCATCTGGTTGGTATTCCTGTCTC 3′ (SEQ ID NO:73) and siRNA #3 sense 5′ AAAATACCAACCAGATGGACCCCTGTCTC 3′ (SEQ ID NO:74) siRNA #4 antisense 5′ AAGCTGGACTCCTCCTACACACCTGTCTC 3′ (SEQ ID NO:75) and siRNA #4 sense 5′ AATGTGTAGGAGGAGTCCAGCCCTGTCTC 3′ (SEQ ID NO:76); siRNA #5 antisense 5′ AAAGTCGACCTTCAGTAAGGACCTGTCTC 3′ (SEQ ID NO:77) and siRNA #5 sense 5′ AATCCTTACTGAAGGTCGACTCCTGTCTC 3′ (SEQ ID NO:78).
  • The Silencer™ siRNA Labeling Kit—FAM (Ambion) was used to label GAPDH siRNA with FAM in order to monitor the uptake of siRNA into RKO and lamina cribrosa cells. After transfection on 8-well culture slides, cells were washed with PBS and fixed for 10 minutes in 3.7% formaldehyde in PBS. The wells were removed and mounting media (Vectashield) was added, followed by a coverslip. Uptake of the FAM-labeled siRNA was visualized under a fluorescent microscope under UV light using a fluorescein filter. The GAPDH siRNA was labeled according to the manufacturer's protocol.
  • Transfection of siRNA
  • RKO cells and lamina cribrosa cells were transfected with siRNA using the same transfection protocol. RKO cells were seeded the day before transfection onto 8-well culture slides or 24-well plates at a density of 46,000 and 105,800 cells per well, respectively. Lamina cribrosa cells were transfected when cell confluence was at 40 to 70% and were generally seeded onto 8-well culture slides at 7500 to 10,000 cells per well three days prior to transfection. Transfection medium sufficient for one well of an 8-well culture slide was prepared by diluting 25.5 pmoles of siRNA stock to a final volume of 21.2 μl in Opti-Mem (Sigma). 0.425 μl of Lipofectamine 2000 was diluted to a final volume of 21.2 μl in Opti-Mem and incubated for 7 to 10 minutes at room temperature. The diluted Lipofectamine 2000 mixture was then added to the diluted siRNA mixture and incubated together at room temperature for 20 to 30 minutes. The cells were washed once with serum-free media before adding 135 μl of serum-free media to the cells and overlaying the 42.4 μl of transfection medium. The cells were placed back in the growth chamber for 4 hours. After the incubation, 65 μl of serum-free media +30% FBS was added to the cells. Transfection of siRNA into cells to be used for Western blot analysis were performed in 24-well plates using the same conditions as the transfections in 8-well slides except that the volumes were increased by 2.3 fold.
  • Following transfection, RKO and lamina cribrosa cells were incubated for 72 hours prior to collection of cellular extract for Western blot analysis. In order to determine the effectiveness of the siRNAs directed against apoptosis-specific eIF-5A to block apoptosis, lamina cribrosa cells were treated with 50 μM of camptothecin (Sigma) and 10 ng/ml of TNF-α (Leinco Technologies) to induce apoptosis either 48 or 72 hours after transfection. The cells were stained with Hoescht either 24 or 48 hours later in order to determine the percentage of cells undergoing apoptosis.
  • Example 9
  • Quantification of HepG2 TNF-α Production
  • HepG2 cells were plated at 20,000 cells per well onto 48-well plates. Seventy two hours later the media was removed and fresh media containing either 2.5 μM control antisense oligonucleotide or 2.5 μM antisense oligonucleotide apoptosis-specific eIF-5A #2 was added to the cells. Fresh media containing antisense oligonucleotides was added after twenty four hours. After a total of 48 hours incubation with the oligonucleotides, the media was replaced with media containing interleukin 1β (IL-1β, 1000 pg/ml; Leinco Technologies) and incubated for 6 hours. The media was collected and frozen (−20° C.) for TNF-α quantification. Additional parallel incubations with untreated cells (without antisense oligonucleotide and IL-1β) and cells treated with only IL-1β were used for controls. All treatments were done in duplicate. TNF-α released into the media was measured by ELISA assays (Assay Designs Inc.) according to the manufacturer's protocol.
  • Example 10
  • The following experiments show that antisense apoptosis-specific eIF-5A nucleotides were able to inhibit expression of apoptosis-specific eIF-5A as well as p53.
  • RKO cells were either left untransfected, mock transfected, or transfected with 200 nM of antisense oligonucleotides apoptosis-specific eIF-5A #1, #2, or #3 (SEQ ID NO: 25, 26, and 27). RKO cells were also transfected with 100 nM of antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26). Forty-eight hours after transfection, the cells were treated with 0.25 μg/ml Actinomycin D. Twenty-four hours later, the cell extract was harvested and 5 μg of protein from each sample was separated on an SDS-PAGE gel, transferred to a PVDF membrane, and Western blotted with an antibody against apoptosis-specific eIF-5A. After chemiluminescent detection, the membrane was stripped and reprobed with an antibody against p53. After chemiluminescent detection, the membrane was stripped again and reprobed with an antibody against actin. FIG. 52 which shows the levels of protein produced by RKO cells after being treated with antisense oligo 1, 2 and 3 (to apoptosis-specific eIF-5a)(SEQ ID NO:25, 26, and 27, respectively). The RKO cells produced less apoptosis-specific eIF-5A as well as less p53 after having been transfected with the antisense apoptosis-specific eIF-5A nucleotides.
  • Example 11
  • The following experiments show that apoptosis-specific eIF-5A nucleotides were able to reduce apoptosis.
  • In one experiment, the lamina cribrosa cell line #506 was either (A) transfected with 100 nM of FITC-labeled antisense oligonucleotide using Oligofectamine transfection reagent or (B) transfected with 10 μM of naked FITC-labeled antisense oligonucleotide diluted directly in serum-free media. After 24 hours fresh media containing 10% FBS and fresh antisense oligonucleotide diluted to 10 μM was added to the cells. The cells, (A) and (B), were fixed after a total of 48 hours and visualized on a fluorescent microscope under UV light using a fluorescein filter. FIG. 53 shows uptake of the flourescently labeled antisense oligonucleotide.
  • In another experiment, the lamina cribrosa cell line #506 was transfected with 10 μM of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of 4 days. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with either 20 μM or 40 μM camptothecin for 48 hours. Antisense oligonucleotide and camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht and TUNEL. See FIG. 54.
  • In another experiment, the lamina cribrosa cell line #506 was transfected with 10 μM of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26). Twenty-four hours later the media was changed and fresh antisense oligonucleotides were added. Forty-eight hours after beginning antisense oligonucleotide treatment, the antisense-oligonucleotides were removed and the cells were treated with 20 μM camptothecin for 3 days. The camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht and TUNEL. See FIG. 55.
  • In yet another experiment, the lamina cribrosa cell line #517 was transfected with 1 μM of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of five days. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with 20 μM camptothecin for either 3 or 4 days. Antisense oligonucleotide and camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht and TUNEL. See FIG. 56.
  • In another experiment, the lamina cribrosa cell line #517 was transfected with 2.5 μM of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of five days. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with 40 μM camptothecin for 3 days. Antisense oligonucleotide and camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 57.
  • In another experiment, the lamina cribrosa cell line #517 was transfected with either 1 μM or 2.5 μM of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of five days. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with 40 μM camptothecin for 3 days. Antisense oligonucleotide and camptothecin-containing media was changed daily. The percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 58.
  • In another experiment, the lamina cribrosa cell line #517 was left either untreated, or was treated with 10 ng/ml TNF-α, 50 μM camptothecin, or 10 ng/ml TNF-α and 50 μM camptothecin. The percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 59.
  • In another experiment, the lamina cribrosa cell lines #506 and #517 were transfected with either 2.5 μM or 5 μM of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of two days. Fresh media containing antisense oligonucleotides was added after 24 hours. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with 50 μM camptothecin and 10 ng/ml TNF-α for 2 days. The percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 60.
  • In another experiment, the lamina cribrosa cell lines #506, #517, and #524 were transfected with 2.5 μM of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 (SEQ ID NO:26) for a total of two days. Fresh media containing antisense oligonucleotides was added after 24 hours. Forty-eight hours after beginning antisense oligonucleotide treatment, the cells were treated with 50 μM camptothecin and 10 ng/ml TNF-α for 2 days. The percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 61.
  • Example 12
  • The following experiments show that cells transfected with siRNAs targeted against apoptosis-specific eIF-5A expressed less apoptosis-specific eIF-5A. The experiments also show that siRNAs targeted against apoptosis-specific eIF-5A were able to reduce apoptosis.
  • In one experiment, the lamina cribrosa cell line #517 was transfected with 100 nM of FAM-labeled siRNA using Lipofectamine 2000 transfection reagent either with serum (A) or without serum (B) during transfection. The cells, (A) and (B), were fixed after a total of 24 hours and visualized on a fluorescent microscope under UV light using a fluorescein filter. See FIG. 62.
  • In another experiment, RKO cells were transfected with 100 nM of siRNA either in the presence or absence of serum during the transfection. Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO:34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-SA (siRNA #1 to #4)(SEQ ID NO:30-33). Seventy-two hours after transfection, the cell extract was harvested and 5 μg of protein from each sample was separated on an SDS-PAGE gel, transferred to a PVDF membrane, and Western blotted with an antibody against apoptosis-specific eIF-5A. After chemiluminescent detection, the membrane was stripped and re-probed with an antibody against bcl-2. After chemiluminescent detection, the membrane was stripped again and re-probed with an antibody against actin. See FIG. 63.
  • In another experiment, lamina Cribrosa cell lines #506 and #517 were transfected with 100 nM of siRNA. Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO:34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-5A (siRNA #1 to #4)(SEQ ID NO:30-33). Seventy-two hours after transfection, the cell extract was harvested and 5 μg of protein from each sample was separated on an SDS-PAGE gel, transferred to a PVDF membrane, and Western blotted with an antibody against apoptosis-specific eIF-5A. After chemiluminescent detection, the membrane was stripped and re-probed with an antibody against actin. See FIG. 64.
  • In another experiment, the lamina cribrosa cell line #506 was transfected with 100 nm of siRNA. Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO: 34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-5A (siRNA #1 to #4)(SEQ ID NO:30-33). Forty-eight hours after transfection, the media was replaced with media containing 50 ,μM camptothecin and 10 ng/ml TNF-α. Twenty-four hours later, the percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 65
  • In another experiment, the lamina cribrosa cell line #506 was transfected with 100 nm of siRNA. Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO:34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-5A (siRNA #1 to #4)(SEQ ID NO:30-33). Seventy-two hours after transfection, the media was replaced with media containing 50 μM camptothecin and 10 ng/ml TNF-α. Twenty-four hours later, the percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 66.
  • In another experiment, the lamina cribrosa cell line #506 was either left untransfected or was transfected with 100 nm of siRNA. Six siRNAs were transfected, two control siRNAs (siRNA #5 (SEQ ID NO:34) and one targeted against GAPDH) and four targeted against apoptosis-specific eIF-5A (siRNA #1 to #4)(SEQ ID NO:30-33). Seventy-two hours after transfection, the media was replaced with media containing 50 μM camptothecin and 10 ng/ml TNF-α. Fresh media was also added to the untransfected, untreated control cells. Forty-eight hours later, the percentage of apoptotic cells was determined by labeling the cells with Hoescht. See FIG. 67.
  • Photographs of Hoescht-stained lamina cribrosa cell line #506 transfected with siRNA and treated with camptothecin and TNF-α from the experiment described in FIG. 67 and example 13 are provided in FIG. 68.
  • Example 13
  • This example shows that treating a human cell line with antisense oligonucleotides directed against apoptosis-specific eIF-5A causes the cells to produce less TNF-α.
  • HepG2 cells were treated with 2.5 μM of either the control antisense oligonucleotide or antisense oligonucleotide apoptosis-specific eIF-5A #2 for a total of two days. Fresh media containing antisense oligonucleotides was added after 24 hours. Additional cells were left untreated for two days. Forty-eight hours after the beginning of treatment, the cells were treated with IL-1β (1000 pg/ml) in fresh media for 6 hours. At the end of the experiment, the media was collected and frozen (−20° C.) for TNF-α quantification. TNF-α released into the media was measured using ELISA assays purchased from Assay Designs Inc. See FIG. 69. Cells that were transfected with antisense oligonucleotides of apoptosis-specific eIF-5A produced less TNF-α.
  • Example 14
  • HT-29 cells (human colon adenocarcinoma) were transfected with either an siRNA against apoptosis-specific eIF-5A or with a control siRNA with the reverse sequence. The siRNA used is as follows:
  • Position 690 (3′UTR) % G/C=48
    5′ AAGCUGGACUCCUCCUACACA 3′ (SEQ ID NO: 79)
  • The control siRNA used is as follows:
  • % G/C=39
    5′ AAACACAUCCUCCUCAGGUCG 3′ (SEQ ID NO: 80)
  • After 48 hours the cells were treated with interferon-gamma (IFN-gamma) for 16 hours. After 16 hours the cells were washed with fresh media and treated with lipopolysaccharide (LPS) for 8 or 24 hours. At each time point (8 or 24 hours) the cell culture media was removed from the cells, frozen, and the TNF-alpha present in the media was quantitated by ELISA. The cell lysate was also harvested, quantitated for protein, and used to adjust the TNF-alpha values to pg/mg protein (to adjust for differences in cell number in different wells). The results of the Western blot and Elisa are provided in FIGS. 74A and B. FIG. 75 are the results of the same experiment except the cells were at a higher density.
  • Example 15
  • Tissue Culture Conditions of U-937 Cell Line
  • U-937 is a human monocyte cell line that grows in suspension and will become adherent and differentiate into macrophages upon stimulation with PMA (ATCC Number CRL-1593.2)(cells not obtained directly from ATCC). Cells were maintained in RPMI 1640 media with 2 mM L-glutamine, 1.5 g/L sodium bicarbonate, 4.5 g/L glucose, 10 mM HEPES, 1.0 mM sodium pyruvate and 10% fetal bovine serum in a 37° C. CO2 (5%) incubator. Cells were split into fresh media (1:4 or 1:5 split ratio) twice a week and the cell density was always kept between 105 and 2×106 cells/ml. Cells were cultured in suspension in tissue culture-treated plastic T-25 flasks and experiments were conducted in 24-well plates.
  • Time Course Experiment
  • Two days before the start of an experiment, the cell density was adjusted to 3×105 cells/ml media. On the day of the experiment, the cells were harvested in log phase. The cell suspension was transferred to 15 ml tubes and centrifuged at 400×g for 10 mins at room temperature. The supernatant was aspirated and the cell pellet was washed/resuspended with fresh media. The cells were again centrifuged at 400×g for 10 mins, the supernatant was aspirated, and the cell pellet was finally resuspended in fresh media. Equal volumes of cell suspension and trypan blue solution (0.4% trypan blue dye in PBS) were mixed and the live cells were counted using a haemocytometer and a microscope. The cells were diluted to 4×105 cells/ml.
  • A 24-well plate was prepared by adding either PMA or DMSO (vehicle control) to each well. 1 ml of cell suspension was added to each well so that each well contained 400,000 cells, 0.1% DMSO ±162 nM PMA. The cells were maintained in a 37° C. CO2 (5%) incubator. Separate wells of cells were harvested at times 0, 24, 48, 72, 96, 99 and 102 h. See FIG. 76 for a summary of the experimental time points and additions.
  • The media was changed at 72 h. Since some cells were adherent and others were in suspension, care was taken to avoid disrupting the adherent cells. The media from each well was carefully transferred into corresponding microcentrifuge tubes and the tubes were centrifuged at 14,000×g for 3 min. The tubes were aspirated, the cell pellets were resuspended in fresh media (1 ml, (−) DMSO, (−) PMA), and returned to their original wells. The cells become quiescent in this fresh media without PMA. At 96 h, LPS (100 ng/ml) was added and cells were harvested at 3 h (99 h) and 6 h (102 h) later.
  • At the time points, the suspension cells and media were transferred from each well into microcentrifuge tubes. The cells were pelleted at 14,000×g for 3 min. The media (supernatant) was transferred to clean tubes and stored (−20° C.) for ELISA/cytokine analysis. The cells remaining in the wells were washed with PBS (1 ml, 37° C.) and this PBS was also used to wash the cell pellets in the corresponding microcentrifuge tubes. The cells were pelleted again at 14,000×g for 3 min. The cells were lysed with boiling lysis buffer (50 mM Tris pH 7.4 and 2% SDS). The adherent cells and the suspension cells from each well were pooled. The samples were boiled and then stored at −20° C.
  • Western Blotting
  • The protein concentration in each cell sample was determined by the BCA (bicinchoninic acid) method using BSA (bovine serum albumin) as the standard protein. Protein samples (5 μg total protein) were separated by 12% SDS-PAGE electrophoresis and transferred to PVDF membranes. The membranes were blocked with polyvinyl alcohol (1 μg/ml, 30 sec) and with 5% skim milk in PBS-t (1 h). The membranes were probed with a mouse monoclonal antibody raised against human eIF-5A (BD Biosciences cat #611976; 1:20,000 in 5% skim milk, 1 h). The membranes were washed 3×10 mins PBS-t. The secondary antibody was a horseradish peroxidase-conjugated antimouse antibody (Sigma, 1:5000 in 1% skim milk, 1 h). The membranes were washed 3×10 mins PBS-t. The protein bands were visualized by chemiluminescence (ECL detection system, Amersham Pharmacia Biotech).
  • To demonstrate that similar amounts of protein were loaded on each gel lane, the membranes were stripped and reprobed for actin. Membranes were stripped (100 mM 2-mercaptoethanol, 2% SDS, 62.5 mM Tris-HCl pH 6.7; 50° C. for 30 mins), washed, and then blocked as above. The membranes were probed with actin primary antibody (actin monoclonal antibody made in mouse; Oncogene, Ab-1; 1:20,000 in 5% skim milk). The secondary antibody, washing, and detection were the same as above.
  • FIG. 77 shows that eIF5A is upregulated during monocyte (U-397) differentiation and subsequent TNF-α secretion.
  • Example 16 Suppression of Il-8 Production in Response to Interferon Gamma by Apoptosis-Specific eIF-5A siRNA
  • HT-29 (human colon adenocarcinoma) cells were transfected with siRNA directed to apoptosis-specific eIF-5A. Approximately 48 hours after transfection the media was changed so that some of the test samples had media with interferon gamma and some of the samples had media without interferon gamma. 16 hours after interferon gamma addition, the cells were washed, and the media, with or without TNF-alpha, was placed on the cells. The media (used for ELISA detection of IL-8) and the cell lysate was harvested 8 or 24 hours later.
  • FIGS. 79 and 80 show that IL-8 is produced in response to TNF-alpha as well as in response to interferon. Priming the cells with interferon gamma prior to TNF treatment causes the cells to produce more IL-8 than either treatment alone. This may be due to the known upregulation of the TNF receptor 1 in response to interferon, so ‘priming’ the cells with interferon allows them to respond to TNF better since the cells have more receptors. siRNA against apoptosis-specific eIF-5A had no effect on IL-8 production in response to TNF alone (previous experiment) however, the siRNA blocked almost all IL-8 produced in response to interferon as well as a significant amount of the IL-8 produced as a result of the combined treatment of interferon and TNF. These results show that the by using siRNAs directed against apoptosis-specific eIF-5A, the inventors have the interferon signaling pathway leading to IL-8, but not the TNF pathway. FIG. 81 is a western showing up-regulation (4 fold at 8 hours) of apoptosis-specific eIF-5A in response to interferon gamma in HT-29 cells.
  • Example 17
  • Human Lamina Cribrosa Culture
  • Paired human eyes were obtained within 48 hours post mortem from the Eye Bank of Canada, Ontario Division. Optic nerve heads (with attached pole) were removed and placed in Dulbecco's modified Eagle's medium (DMEM) supplemented with antibiotic/antimycotic, glutamine, and 10% FBS for 3 hours. The optic nerve head (ONH) button was retrieved from each tissue sample and minced with fine dissecting scissors into four small pieces. Explants were cultured in 12.5 cm2 plastic culture flasks in DMEM medium. Growth was observed within one month in viable explants. Once the cells reached 90% confluence, they were trypsinized and subjected to differential subculturing to produce lamina cribrosa (LC) and astrocyte cell populations. LC cells were enriched by subculture in 25 cm2 flasks in DMEM supplemented with gentamycin, glutamine, and 10% FBS. Cells were maintained and subcultured as per this protocol.
  • The identity and population purity of cells populations obtained by differential subculturing was characterized using differential fluorescent antibody staining on 8 well culture slides. Cells were fixed in 10% formalin solution and washed three times with Dulbecco's Phosphate Buffered Saline (DPBS). Following blocking with 2% nonfat milk in DPBS, antibodies were diluted in 1% BSA in DPBS and applied to the cells in 6 of the wells. The remaining two wells were treated with only 1% bovine serum albumin (BSA) solution and only secondary antibody as controls. Cells were incubated with the primary antibodies for one hour at room temperature and then washed three times with DPBS. Appropriate secondary antibodies were diluted in 1% BSA in DPBS, added to each well and incubated for 1 hour. Following washing with DPBS, the slide was washed in water, air-dried, and overlayed with Fluoromount (Vector Laboratories). Immunofluorescent staining was viewed under a fluorescent microscope with appropriate filters and compared to the control wells that were not treated with primary antibody. All primary antibodies were obtained from Sigma unless otherwise stated. All secondary antibodies were purchased from Molecular Probes. Primary antibodies used to identify LC cells were: anti-collagen I, anti-collagen IV, anti-laminin, anti-cellular fibronectin, anti-glial fibrillary acidic protein (GFAP), and anti-alpha-smooth muscle actin. Cell populations were determined to be comprised of LC cells if they stained positively for collagen I, collagen IV, laminin, cellular fibronectin, alpha smooth muscle actin and negatively for glial fibrillary (GFAP). In this study, two sets of human eyes were used to initiate cultures. LC cell lines #506 and #517 were established from the optic nerve heads of and 83-year old male and a 17-year old male, respectively. All LC cell lines have been fully characterized and found to contain greater than 90% LC cells.
  • Treatment of LC cells
  • Apoptosis was induced in lamina cribrosa cells using a combination of 50 μM camptothecin (Sigma) and 10 ng/ml TNF-α (Leinco Technologies). The combination of camptothecin and TNF-α was found to be more effective at inducing apoptosis than either camptothecin or TNF-α alone.
  • Construction and Transfection of siRNAs
  • Small inhibitory RNAs (siRNAs) directed against human apoptosis-specific eIF-5A were used to specifically suppress expression of eIF5A in lamina cribrosa cells. Six siRNAs were generated by in vitro transcription using the Silencer™ siRNA Construction Kit (Ambion Inc.). Four siRNAs were generated against human apoptosis-specific eIF-5A (siRNAs #1 to #4). Two siRNAs were used as controls; an siRNA directed against GAPDH provided in the kit, and an siRNA (siRNA #5), which had the reverse sequence of the apoptosis-specific eIF-5A specific siRNA #1, but does not itself target eIF5A. The siRNAs were generated according to the manufacturer's protocol. The eIF5A and control siRNA targets had the following sequences:
    siRNA # 1 5′
    AAAGGAATGACTTCCAGCTGA 3′; (SEQ ID NO: 81)
    siRNA # 2 5′
    AAGATCGTCGAGATGTCTACT 3′; (SEQ ID NO: 82)
    siRNA # 3 5′
    AAGGTCCATCTGGTTGGTATT 3′; (SEQ ID NO: 83)
    siRNA # 4 5′
    AAGCTGGACTCCTCCTACACA 3′; (SEQ ID NO: 84)
    siRNA # 5′
    AAAGTCGACCTTCAGTAAGGA 3′. (SEQ ID NO: 85)

    Lamina cribrosa cells were transfected with siRNA using LipofectAMINE 2000.
  • Lamina cribrosa cells were transfected when cell confluence was at 40 to 70% and were generally seeded onto 8-well culture slides at 7500 cells per well three days prior to transfection. Transfection medium sufficient for one well of an 8-well culture slide was prepared by diluting 25.5 pmoles of siRNA to a final volume of 21.2 μl in Opti-Mem (Sigma). 0.425 μl of Lipofectamine 2000 was diluted to a final volume of 21.2 μl in Opti-Mem and incubated for 7 to 10 minutes at room temperature. The diluted Lipofectamine 2000 mixture was then added to the diluted siRNA mixture and incubated together at room temperature for 20 to 30 minutes. The cells were washed once with serum-free media before adding 135 μl of serum-free media to the cells and overlaying 42.4 μl of transfection medium. The cells were placed back in the growth chamber for 4 hours. After the incubation, 65 μl of serum-free media plus 30% FBS was added to the cells. Transfection of siRNA into cells to be used for Western blot analysis were performed in 24-well plates using the same conditions as the transfections in 8-well slides except that the volumes were increased by 2.3 fold. Following transfection, lamina cribrosa cells were incubated for 72 hours prior to treatment with 50 μM of camptothecin (Sigma) and 10 ng/ml of TNF-α (Leinco Technologies) to induce apoptosis. Cell lysates were then harvested for Western blotting or the cells were examined for apoptosis
  • Detection of Apoptotic Cells
  • Transfected cells that had been treated with TNF-α and camptothecin for 24 hours were stained with Hoescht 33258 in order to determine the percentage of cells undergoing apoptosis. Briefly, cells were fixed with a 3:1 mixture of absolute methanol and glacial acetic acid and then incubated with Hoescht stain (0.5 pg/ml Hoescht 33258 in PBS). After a 10 minute incubation in the dark, the staining solution was discarded, the chambers separating the wells of the culture slide were removed, and the slide was washed 3 times for 1 minute with deionized water. After washing, a few drops of Mcllvaine's buffer (0.021 M citric acid, 0.058 M Na2HPO4.7H2O; pH 5.6) was added to the cells and overlaid with a coverslip. The stained cells were viewed under a fluorescent microscope using a UV filter. Cells with brightly stained or fragmented nuclei were scored as apoptotic. A minimum of 200 cells were counted per well. The DeadEnd™ Fluorometric TUNEL (Promega) was also used to detect the DNA fragmentation that is a characteristic feature of apoptotic cells. Following Hoescht staining, the culture slide was washed briefly with distilled water, and further washed by immersing the slide twice for 5 minutes in PBS (137 mM NaCl, 2.68 mM KCl, 1.47 mM KH2PO4, 8.1 mM Na2HPO4), blotting the slide on paper towel between washes. The cells were permeabilized by immersing them in 0.2% Triton X-100 in PBS for 5 minutes. The cells were then washed again by immersing the slide twice for 5 minutes in PBS and blotting the slide on paper towel between washes. 25 μl of equilibration buffer [200 mM potassium cacodylate (pH 6.6), 25 mM Tris-HCl (pH 6.6), 0.2 mM dithiothreitol, 0.25 mg/ml bovine serum albumin, and 2.5 mM cobalt chloride] was added per well and incubated for 5 to 10 minutes. During equilibration, 30 μl of reaction mixture was prepared for each well by mixing in a ratio of 45:5:1, respectively, equilibration buffer, nucleotide mix [50 μM fluorescein-12-dUTP, 100 μM dATP, 10 mM Tris-HCl (pH 7.6), and 1 mM EDTA], and terminal deoxynucleotidyl transferase enzyme (Tdt, 25 U/μl). After the incubation in equilibration buffer, 30 μl of reaction mixture was added per well and overlayed with a coverslip. The reaction was allowed to proceed in the dark at 37° C. for 1 hour. The reaction was terminated by immersing the slide in 2×SSC [0.3 M NaCl, and 30 mM sodium citrate (pH 7.0)] and incubating for 15 minutes. The slide was then washed by immersion in PBS three times for 5 minutes. The PBS was removed by sponging around the wells with a Kim wipe, a drop of mounting media (Oncogene research project, JA1750-4ML) was added to each well, and the slide was overlayed with a coverslip. The cells were viewed under a fluorescent microscope using a UV filter (UV-G 365, filter set 487902) in order to count the Hoescht-stained nuclei. Any cells with brightly stained or fragmented nuclei were scored as apoptotic. Using the same field of view, the cells were then viewed using a fluorescein filter (Green H546, filter set 48915) and any nuclei fluorescing bright green were scored as apoptotic. The percentage of apoptotic cells in the field of view was calculated by dividing the number of bright green nuclei counted using the fluorescein filter by the total number of nuclei counted under the UV filter. A minimum of 200 cells were counted per well.
  • Protein Extraction and Western Blot Analysis
  • Protein was isolated for Western blotting from lamina cribrosa cells growing on 24-well plates by washing the cells twice in PBS (8 g/L NaCl, 0.2 g/L KCl, 1.44 g/L Na2HPO4, and 0.24 g/L KH2PO4) and then adding 50 μl of lysis buffer [2% SDS, 50 mM Tris-HCl (pH 7.4)]. The cell lysate was collected in a microcentrifuge tube, boiled for 5 minutes and stored at −20° C. until ready for use. Protein concentrations were determined using the Bicinchoninic Acid Kit (BCA; Sigma). For Western blotting, 5 μg of total protein was separated on a 12% SDS-polyacrylamide gel. The separated proteins were transferred to a polyvinylidene difluoride membrane. The membrane was then incubated for one hour in blocking solution (5% skim milk powder, 0.02% sodium azide in PBS) and washed three times for 15 minutes in PBS-T (PBS+0.05% Tween-20). The membrane was stored overnight in PBS-T at 4° C. After being warmed to room temperature the next day, the membrane was blocked for 30 seconds in 1 μg/ml polyvinyl alcohol. The membrane was rinsed 5 times in deionized water and then blocked for 30 minutes in a solution of 5% milk in PBS. The primary antibody was preincubated for 30 minutes in a solution of 5% milk in PBS prior to incubation with the membrane. The primary antibodies used were anti-eIF5A (BD Transduction Laboratories) at 1:20,000 and anti-β-actin (Oncogene). The membranes were washed three times in PBS-T and incubated for 1 hour with the appropriate HRP-conjugated secondary antibodies diluted in 1% milk in PBS. The blot was washed and the ECL Plus Western blotting detection kit (Amersham Pharmacia Biotech) was used to detect the peroxidase-conjugated bound antibodies.
  • Results
  • Two lamina cribrosa (LC) cell lines were established from optic nerve heads obtained from male donors ranging in age from 83 years (#506) to 17 years (#517). The cells isolated from the human lamina cribrosa had the same broad, flat morphology with prominent nucleus observed in other studies (Lambert et al., 2001). Consistent with the characterizations of other groups, the LC cells showed immunoreactivity to alpha smooth muscle actin (FIG. 82 a) as well as to a number of extracellular matrix proteins including cellular fibronectin (FIG. 82 b), laminin (FIG. 82 c), collagen I, and collagen IV (data not shown) (Clark et al., 1995; Hernandez et al., 1998; Hernandez and Yang, 2000; Lambert et al.; 2001). Negative immunoreactivity of the LC cells to glial fibrillary acidic protein (GFAP) was also observed consistent with previous findings (FIG. 82 d) (Lambert et al., 2001). These findings support the identification of the isolated cells as being LC cells rather than optic nerve head astrocytes.
  • Since TNF-α is believed to play an important role during the glaucomatous process, the susceptibility of LC cells to the cytotoxic effects of TNF-α was examined. Confluent LC cells were exposed to either camptothecin, TNF-α, or a combination of camptothecin and TNF-α for 48 hours (FIG. 83). Hoescht staining revealed that TNF-α alone was not cytotoxic to LC cells. Treatment with camptothecin resulted in approximately 30% cell death of the LC cells. However, a synergistic increase in apoptosis was observed when LC cells were treated with both camptothecin and TNF-α, a treatment which resulted in the death of 45% of LC cells by 48 hours. These results indicate that LC cells are capable of responding to the cytotoxic effects of TNF-α when primed for apoptosis by camptothecin.
  • EIF5A is a nucleocytoplasmic shuttle protein known to be necessary for cell division and recently suggested to also be involved during apoptosis. The expression of apoptosis-specific eIF-5A protein in LC cells being induced to undergo apoptosis by either camptothecin, or camptothecin plus TNF-α. The expression of apoptosis-specific eIF-5A did not alter significantly upon treatment with camptothecin except perhaps to decrease slightly (FIG. 84A). However, a significant upregulation of apoptosis-specific eIF-5A protein was observed after 8 and 24 hours of camptothecin plus TNF-α treatment (FIG. 84B). These results indicate that of apoptosis-specific eIF-5A expression is induced specifically by exposure TNF-α and expression correlates to the induction of apoptosis. This points to a role for apoptosis-specific eIF-5A in the apoptotic pathway downstream of TNF-α receptor binding.
  • In order to examine the importance of apoptosis-specific eIF-5A expression during TNF-α-induced apoptosis in LC cells, a series of four siRNAs (siRNAs #1 to #4) targeting apoptosis-specific eIF-5A were designed and synthesized by in vitro transcription. To determine the effectiveness of the siRNAs in suppressing apoptosis-specific eIF-5A protein expression, LC cell lines #506 and #517 were transfected with each of the siRNAs and expression of apoptosis-specific eIF-5A protein in the cell lysate was examined 72 hours later (FIG. 85). For comparison, cells were also transfected with either an siRNA against GAPDH and/or a control siRNA (siRNA #5) having the same chemical composition as siRNA #1 but which does not recognize apoptosis-specific eIF-5A. All siRNAs directed against apoptosis-specific eIF-5A were capable of significantly suppressing apoptosis-specific eIF-5A expression in both LC cell lines (FIG. 85). The GAPDH siRNA was used as an additional control because, unlike the control siRNA #5 which simply has the reverse sequence of siRNA #1 and does not have a cellular target, it is an active siRNA capable of suppressing the expression of it's target protein, GAPDH (data not shown). All four siRNAs against apoptosis-specific eIF-5A were also capable of protecting transfected LC cells (#506) from apoptosis induced by 24 hour treatment with TNF-α and camptothecin (FIG. 86). Using Hoescht staining to detect cell death, the siRNAs (siRNAs #1 to #4) were found to be able to reduce apoptosis of LC cells by 59% (siRNA #1), 35% (siRNA #2), 50% (siRNA #3), and 69% (siRNA #4). Interestingly, the siRNA against GAPDH was also able to reduce apoptosis of LC cells by 42% (FIG. 86). GAPDH is known to have cellular functions outside of it's role as a glycolytic enzyme, including a proposed function during apoptosis of cerebellar neurons (Ishitani and Chuang, 1996; Ishitani et al., 1996a; Ishitani et al., 1996b). In a similar experiment we also demonstrated that siRNA #1 was able to reduce apoptosis of the LC line #517 by 53% in response to TNF-α and camptothecin indicating that apoptosis-specific eIF-5A siRNAs are protective for LC cells isolated from different optic nerve heads (FIG. 87). These results indicate that apoptosis-specific eIF-5A does have a function during apoptosis and may be an important intermediate in the pathway leading to TNF-α-induced apoptosis in LC cells.
  • In order to confirm that LC cells exposed to TNF-a and camptothecin were dying by classical apoptosis, DNA fragmentation was evaluated in situ using the terminal deoxynucleotidyl transferase-mediated dUTP-digoxigenin nick end labeling (TUNEL) method. LC cells (#506) were treated with TNF-α and camptothecin for 24 hours, 3 days after transfection with either an apoptosis-specific eIF-5A siRNA (siRNA #1) or a control siRNA (siRNA #5). The cells were also stained with Hoescht to facilitate visualization of the nuclei. 46% of LC cells transfected with the control siRNA were positive for TUNEL staining while only 8% of LC cells transfected with apoptosis-specific eIF-5A siRNA #1 were positively labeled indicating that the apoptosis-specific eIF-5A siRNA provided greater than 80% protection from apoptosis (FIG. 88). Similar results were obtained with apoptosis-specific eIF-5A siRNA #4 which provided greater than 60% protection from apoptosis relative to the control siRNA (data not shown).
  • Example 18
  • Blood Collection and Preparation of PBMCs
  • Approximately 10 ml of blood was collected from each healthy donor. The blood was collected by venapuncture in a vacutainer containing sodium citrate as the anti-coagulant. The samples were processed within 24 hours of collection.
  • A 60% SIP (9 parts v/v Percoll with 1 part v/v 1.5M NaCl) was cushioned on the bottom of 15 ml conical tubes. The blood was then layered overtop with minimal mixing of the blood and Percoll cushion. The samples were centrifuged for 30 minutes total at 100×g with slow acceleration in the first 5 minutes and slow deceleration in the last 5 minutes. The pure serum at the very top of the resulting gradient was removed and the white cushion (1-2 ml) of PBMCs was collected and added dropwise to a tube containing 10 ml of warm RPMI plus 15% FBS. The PBMCs were pelleted and counted.
  • Stimulation to Induce Cytokine Production in PBMCs Over a Time Course
  • PBMCs were isolated and seeded at 2×105 to 5×105 cells/well. The cells were treated with phorbol 12-myristate 13-acetate (PMA; 100 ng/well). At 72 hours the media was replaced and did not contain any stimulating factors. Then at 96 hours after PMA addition to PBMCs, lipopolysaccharide (LPS; 100 ng/well; from E. coli, serotype 0111) was added to the wells. Samples were collected before LPS addition (96 h), and at various times after addition as outlined in FIG. 91. Both adherent cells (likely to be monocytes and macrophages) were collected with the floating cells (likely to be lymphocytes). To collect samples for analysis of cytokine secretion, the media from each well was transferred to clean microcentrifuge tubes and cleared of any debris by centrifugation at 13000×g for 3 minutes. The resulting pellet was collected with the adherent cells. The media was stored at −20° C. in 200-250 μl aliquots prior to analysis. The cells were washed with 1 ml of 37° C. phosphate buffered saline (PBS) and then lysed in boiling lysis buffer (50 mM Tris pH 7, 2% SDS; 100 μl per well). The cell lysates were boiled and stored frozen at −20° C. The Western blot is shown in FIG. 92 and the corresponding ELISA in FIG. 93.
  • PBMC Stimulation to Induce Apoptosis-Specific eIF-5A Expression
  • PBMCs were collected and seeded at 2×105 to 5×105 cells/well. To determine which stimulators induce apoptosis-specific eIF-5A, as well as to see if they act synergistically, the PBMCs were stimulated with phytohemagglutinin (PHA; 100 ng/ml), phorbol 12-myristate 13-acetate (PMA; 100 ng/ml), lipopolysaccharide (LPS; 100 ng/ml) or all three (each at 100 ng/ml). The samples were collected 12 and 36 hours after stimulation and analyzed for apoptosis-specific eIF-5A expression (FIG. 94).
  • Transfection of PBMCs
  • PBMCs were transfected the day they were prepared. Cells were seeded at 2×10 5 to 5×105 cells/well (150 μl per well for transfection in a 24-well plate). They were either transfected individually in each well ( Donors 77, 78 and 79; FIGS. 95 and 96) or all at once in a conical tube before seeding ( Donors 80 and 84; FIG. 96). For each well of cells to be transfected, 15 pmoles of siRNA was diluted in 50 μl of Opti-MEM (Sigma). 1 μl of Lipofectamine 2000 (Invitrogen) was diluted in 49 μl of Opti-MEM, incubated for 7 to 10 minutes, added to the diluted siRNA and incubated 25 minutes. The transfection medium was overlayed onto the cells were placed in the 37° C. growth chamber for 4 hours. The final transfection medium contained 9% serum. After the incubation, 250 μl of serum-free RPMI+21% FBS was added to the cells to make the final serum concentration 15%.
  • Stimulation to Induce Cytokine Production in PBMCs Post Transfection
  • 72 hours after transfection of the PBMCs, as outlined above, lipopolysaccharide (LPS; 100 ng/well; from E. coli, serotype 0111) was added to the cells in 500 μl of media. The samples were collected at 24 hours post stimulation. Both wells that were treated with LPS and wells that were transfected only (i.e. no stimulation) were collected. To collect samples for analysis of cytokine secretion, the media from each well was transferred to clean microcentrifuge tubes and cleared of any debris by centrifugation at 13000×g for 3 minutes. The resulting pellet was collected with the adherent cells. The media was stored at −20° C. in 200-250 μl aliquots prior to analysis. The cells were washed with 1 ml of 37° C. phosphate buffered saline (PBS) and then lysed in boiling lysis buffer (50 mM Tris pH 7, 2% SDS; 100 μl per well). The cell lysates were boiled and stored frozen at −20° C. for BCA protein quantitation.
  • Example 19
  • Cell Culture
  • HT-29, a human colorectal adenocarcinoma cell line, was maintained in RPMI with 10% fetal bovine serum (FBS). U937, a histiocytic lymphoma cell line, was grown in suspension in RPMI with 10% FBS. Both cell lines were maintained in a humidified environment at 37° C. and 5% CO2. For experiments with U937 cells, cells were counted and adjusted to 3×105 cells/ml two days before the start of the experiment. On the first day of the experiment, cells were collected by centrifugation at 400×g for 10 mins, the cell pellet was resuspended in fresh RPMI media with 10% FBS, the centrifugation was repeated, and the repelleted cells were resuspended in fresh RPMI media without FBS. The cells were counted and adjusted to 2×106 cells/ml.
  • siRNA
  • siRNA sequences were designed based on the human apoptosis-specific eIF-5A sequence and were synthesized by Dharmacon RNA Technologies. The apoptosis-specific eIF-5A siRNA (h5A1) target sequence was: 5′ NNGCUGGACUCCUCCUACACA 3′. The corresponding double stranded siRNA sequence was:
       5′ GCUGGACUCCUCCUACACAdTdT 3′
    3′ dTdTCGACCUGAGGAGGAUGUGU 5′
  • The control siRNA (hcontrol) sequence was 5′ NNACACAUCCUCCUCAGGUCG 3′. The corresponding double stranded siRNA sequence was:
         5′ ACACAUCCUCCUCAGGUCGdTdT 3′
    3′ dTdTUGUGUAGGAGGAGUCCAGC 5′

    Transfection of HT-29 Cells
  • The day before transfection, HT-29 cells were seeded at 105,000 cells per well onto a 24-well plate. For each well of cells to be transfected, 25.5 pmoles of siRNA was diluted in 50 μl of Opti-Mem (Sigma). 1 μl of Lipofectamine 2000 (Invitrogen) was diluted in 49 μl of Opti-Mem, incubated for 7 to 10 minutes and added to the diluted siRNA and incubated 25 minutes. The cells to be transfected were washed once with serum-free RPMI before adding 300 μl of serum-free RPMI and overlaying 100 μl of transfection medium. The cells were placed back in the growth chamber for 4 hours. After the incubation, 300 μl of serum-free RPMI+30% FBS was added to the cells.
  • Electroporation of U937 Cells
  • apoptosis-specific eIF-5A and control siRNA were diluted in Opti-Mem media (Sigma). 400 μl cells (800,000 cells) and 100 pmoles siRNA were mixed in a 0.4 mm electroporation cuvette. The cells were electroporated at 300 V, 10 mSec, 1 pulse with an ECM 830 Electrosquare porator (BTX, San Diego, Calif.). Following electroporation, the cells were gently mixed and added to wells containing RPMI and concentrated FBS so that the final FBS concentration was 10%.
  • Treatment of HT-29 Cells
  • TNF-α production was induced in HT-29 cells according to the method developed by Suzuki et al. 2003. HT-29 cells were primed with 200 units/ml interferon gamma (Roche Diagnostics) 48 hours after transfection. After 16 hours of interferon gamma (IFNγ) priming the cells were washed with media and lipopolysaccharide (LPS; 100 ng/ml; from E. coli, serotype 0111; Sigma) was added at 100 μg/ml. After 8 or 24 hours of LPS stimulation, the media from each well was transferred to microcentrifuge tubes and stored at −20° C. until assayed for TNFα by ELISA. The cells were washed with 1 ml of phosphate buffered saline (PBS) heated to 37° C. and then lysed in boiling lysis buffer (50 mM Tris pH 7, 2% SDS). The cell lysates were boiled and stored frozen at −20° C. The protein concentration in the cell lysates was determined by bicinchoninic acid assays (BCA) with bovine serum albumin used as the standard.
  • IL-8 production was induced in HT-29 cells by treatment with IFNγ. HT-29 cells were treated with 200 units/ml IFNγ 48 hours after transfection. After 24 hours of treatment, the media from each well was transferred to microcentrifuge tubes and stored at −20° C. until assayed for IL-8 by liquid-phase electrochemiluminescence (ECL). The cells were washed with 1 ml of phosphate buffered saline (PBS) heated to 37° C. and then lysed in boiling lysis buffer (50 mM Tris pH 7, 2% SDS). The cell lysates were boiled and stored frozen at −20° C. The protein concentration in the cell lysates was determined by bicinchoninic acid assays (BCA) with bovine serum albumin used as the standard.
  • Induction of Differentiation in U937 Cells
  • U937 cells were collected and counted 16 hours after electroporation. 200,000 cells in 1 ml of media were added to each well of 24-well plates. Macrophage differentiation was stimulated by adding phorbol 12-myristate 13-acetate (PMA; 100 ng/ml). After 48 h with PMA, >80% of the monocytes had transformed from cells in suspension (monocytes) to adherent cells (macrophages). At 48 hours the media and any non-adherent cells were removed and fresh RPMI media with 10% FBS (1 ml per well) was added. The cells were left for 24 hours in fresh media to become quiescent.
  • Stimulation to Induce Cytokine Production in U937 Cells
  • 72 hours after PMA addition to U937 cells, lipopolysaccharide (LPS; 100 ng/ml; from E. coli, serotype 0111), interferonγ (IFNγ; 100 Units/ml), or a combination of LPS and IFNγ were added to the wells. Samples were collected before stimulator addition (72 h), and at various times after addition as outlined in FIG. 108. To collect samples for analysis of cytokine secretion, the media from each well was transferred to clean microcentrifuge tubes and cleared of any debris by centrifugation at 13000×g for 3 mins. The media was stored at −20° C. in 200-250 ul aliquots prior to analysis. The cells were washed with 1 ml of 37° C. phosphate buffered saline (PBS) and then lysed in boiling lysis buffer (50 mM Tris pH 7, 2% SDS; 75 μl per well). Like wells were pooled. The cell lysates were boiled and stored frozen at −20° C.
  • Cytokine Quantification
  • All media samples were stored frozen at −20° C. TNFα was quantified using ELISA kits from Assay Designs according to the manufacturer's instructions with supplied standards for 0-250 pg TNFα/ml. For U937 experiments media samples for TNFα were diluted 20 fold (0 h, 3 h LPS) or 80 fold (6 h, 24 h, 30 h LPS) with RPMI+10% FBS. IL-1β, IL-8, and IL-6 were quantified by liquid-phase electrochemiluminescence (ECL). Media from HT-29 experiments were not diluted. All cytokine measurement results were corrected for the amount of total cellular protein (mg) per well.
  • IL-8, IL-1β, and IL-6 were assayed by liquid-phase. Briefly, a purified monoclonal mouse anti- mouse anti-human IL-8, IL-6 or IL-1β (R & D Systems) were labeled with biotin (Igen, Inc., Gaithersburg, Md.). In addition, the goat anti-human IL-8, IL-6, or IL-1β antibody (R & D) were labeled with ruthenium (Igen) according to the manufacturer's instructions. The biotinylated antibodies were diluted to a final concentration of 1 mg/mL in PBS, pH 7.4, containing 0.25% BSA, 0.5% Tween-20 and 0.01% azide, (ECL buffer). Per assay tube, 25 mL of the biotinylated antibodies were pre-incubated at room temperature with 25 mL of a 1 mg/mL solution of streptavidin-coated paramagnetic beads (Dynal Corp., Lake Success, N.Y.) for 30 min by vigorous shaking. Samples to be tested (25 mL) which had been diluted in RPMI or standards were added to tubes followed by 25 mL of ruthenylated antibody (final concentration 1 mg/mL, diluted in ECL buffer). The tubes were then shaken for an additional 2 hours. The reaction was quenched by the addition of 200 mL/tube of PBS and the amount of chemiluminiscence determined using an Origen Analyzer (Igen).
  • SDS-PAGE and Western Blotting
  • The protein concentration in the cell lysates was determined by bicinchoninic acid assays (BCA) with bovine serum albumin used as the standard. 5 μg of total cellular protein was separated by either 10% or 14% SDS-PAGE (sodium dodecyl sulfate polyacrylamide gel electrophoresis). 10% gels were used for analysis of proteins above 50 kDa (TLR4, IFNγ, TNF-R1, iNOS) while 14% gels were used for apoptosis-specific eIF-5A (17 kDa). Gels were transferred to polyvinylidene fluoride (PVDF) membranes with transfer buffer (48 mM Tris, 39 mM glycine, 1.3 mM SDS, pH 9.2; 15V for 18 mins) using a semi-dry transfer unit (Bio-Rad). Membranes were blocked for 1 hour with 5% skim milk in PBS-t (PBS with 0.1% Tween 20). Primary antibodies were diluted in the blocking solution and all blots were incubated at room temperature with shaking. Primary antibodies used were apoptosis-specific eIF-5A (BD Biosciences; 1:20,000; incubate 1 hour; recognizes both apoptosis-specific eIF-5A and eIF5A2), TLR4 (Santa Cruz Biotechnology Inc; TLR4 (H-80): sc-10741; 1:1000; incubate 2 hours), IFN-γRα (Santa Cruz Biotechnology Inc; IFN-γRα (C-20): sc-700; 1:1000; incubate 1 hour), TNF-R1 (Santa Cruz Biotechnology Inc; TNF-R1 (H-5): sc-8436; 1:200; incubate 3 hours), iNOS (BD Transduction Laboratories: 610431; 1:10,000; incubate 1 hour) and β-actin (Oncogene; actin (Ab-1); 1:20,000; incubate 1 hour). Following primary antibody incubations, blots were washed 3 times for 5-10 minutes with PBS-t. Horseradish peroxidase-conjugated (HRP) secondary antibodies were diluted in 1% skim milk and incubated with the membrane for 1 hour. Secondary antibodies used were anti-mouse IgG-HRP (Sigma; 1:5000; for apoptosis-specific eIF-5A and TNF-R1), anti-rabbit IgG-HRP (Amersham Pharmacia Biotech; 1:2500; for TLR4 and IFNγ-Rα), anti-mouse IgM-HRP (Calbiochem; 1:5000; for actin). Following secondary antibody incubations, blots were washed 4 times for 5-10 mins with PBS-t. Blots were developed with enhanced chemiluminescent detection reagent (ECL; Amersham Pharmacia Biotech) according to the manufacturers instructions and bands were visualized on X-ray film (Fuji).
  • RT-PCR
  • RT-PCR was performed according to Medvedev et al. 2002 in order to observe changes in TLR4 mRNA expression in transfected HT-29 cells in response to IFNγ. Expression of GAPDH was used as a control to show that equal amounts of cDNA were being used between samples. Increasing PCR cycles (20, 25, 30, and 35) were used to determine the optimal cycle number that resulted in detectable amplified products under nonsaturating conditions. PCR products were detected by ethidium bromide-incorporation and were separated by agarose gel electrophoresis. RT-PCR of total MRNA isolated from siRNA-transfected HT-29 cells treated with or without IFNγ for 6 hours was used to detect TLR4 and GAPDH transcripts. HT-29 cells were transfected with siRNA as described above. 48 hours after transfection, the cells were treated with 200 units/ml IFNγ. Control cells which were not treated with IFNγ received only a media change. Total mRNA was isolated using the GenElute Mammalian RNA miniprep kit (Sigma) according to the manufacturer's protocol for adherent cells. The media was removed and the cells were washed twice with warm PBS. Lysis buffer was added to the cells and the lysate was transferred to a microcentrifuge tube and total RNA was isolated according to the manufacturer's protocol.
    The primers for TLR4 (NM_003266) were:
    Forward 5′ CGGATGGCAACATTTAGAATTAGT 3′
    Reverse 5′ TGATTGAGACTGTAATCAAGAACC 3′
    Expected fragment size: 674 bp
    The primers for GAPDH (BC023632) were:
    Forward 5′ CTGATGCCCCCATGTTCGTCAT 3′
    Reverse 5′ CCACCACCCTGTTGCTGTAG 3′
    Expected fragment size: 599 bp
  • The total RNA was reverse transcribed using the following conditions:
    Mix:
    RNA 2.5 μg
    Poly (T) primer 6.25 μl
    Depc water to 13.75 μl
    Heat 70° C. 5 min
    Chill on ice 5 min
    Add:
    5× AMV Buffer 5.0 μl
    dNTPs (10 mM) 2.5 μl
    Rnase Inhibitor 1.25 μl
    AMV RT 2.5 μl
    Heat
    42° C. 60 min
    Heat
    70° C. 10 min
  • A single PCR reaction was performed using the following conditions:
    10× Tsg buffer 2.0 μl
    dNTP (10 mM) 0.4 μl
    forward primer (25 pmol/μl) 0.4 μl
    reverse primer (25 pmol/μl) 0.4 μl
    MgCl2 (15 mM) 2.0 μl
    cDNA 0.8 μl
    H2O 13.88 μl
    Tsg polymerase 0.12 μl
    The PCR conditions for TLR4 were:
    Heat to 95° C. 5 min
    20, 25, 30, or 35 cycles of: 95° C. 1 min
    55° C. 1 min
    72° C. 2 min
    Extend at 72° C. for 10 min
    Sink to 4° C.
    The PCR conditions for GAPDH were:
    Heat to 95° C. 5 min
    20, 25, 30, or 35 cycles of: 95° C. 1 min
    57° C. 1 min
    72° C. 2 min
    Extend at 72° C. for 10 min
    Sink to 4° C.

Claims (28)

1. An antisense oligonucleotide of apoptosis-specific eIF-5A wherein said antisense oligonucleotide suppresses endogenous expression of apoptosis-specific eIF-5A in a cell.
2. The antisense oligonucleotide of claim 1, wherein apoptosis-specific eIF-5A is encoded by nucleotide sequence of SEQ ID NO:20.
3. The antisense oligonucleotide of claim 1, wherein apoptosis-specific eIF-5A is encoded by nucleotide sequence of SEQ ID NO:41.
4. The antisense oligonucleotide of claim 1 wherein the antisense oligonucleotide has the sequence set forth in SEQ ID NO:35.
5. The antisense oligonucleotide of claim 1 wherein the antisense oligonucleotide has the sequence set forth in SEQ ID NO:37.
6. The antisense oligonucleotide of claim 1 wherein the antisense oligonucleotide has the sequence set forth in SEQ ID NO:39.
7. An expression vector for the transfection of a mammalian cell comprising the antisense oligonucleotide of claim 1 and regulatory sequences operatively linked to the antisense oligonucleotide to allow transcription of said antisense oligonucleotide in said cell.
8. An expression vector for the transfection of a mammalian cell comprising the antisense oligonucleotide of claim 4 and regulatory sequences operatively linked to the antisense oligonucleotide to allow transcription of said antisense oligonucleotide in said cell.
9. An expression vector for the transfection of a mammalian cell comprising the antisense oligonucleotide of claim 5 and regulatory sequences operatively linked to the antisense oligonucleotide to allow transcription of said antisense oligonucleotide in said cell.
10. An expression vector for the transfection of a mammalian cell comprising the antisense oligonucleotide of claim 6 and regulatory sequences operatively linked to the antisense oligonucleotide to allow transcription of said antisense oligonucleotide in said cell.
11. A method of inhibiting expression of apoptosis-specific eIF-5A in a cell, the method comprising administering the expression vector of claim 7 to said cell whereby said apoptosis-specific eIF-5A antisense oligonucleotide inhibits expression of endogenous apoptosis-specific eIF-5A in said cell.
12. A method of inhibiting expression of apoptosis-specific eIF-5A in a cell, the method comprising administering the expression vector of claim 8, 9, or 10 to said cell whereby said apoptosis-specific eIF-5A antisense oligonucleotide inhibits expression of endogenous apoptosis-specific eIF-5A in said cell.
13. The method of claim 11 wherein said inhibition of expression of endogenous apoptosis-specific eIF-5A in said cell has an effect on the cell selected from the group consisting of suppressing apoptosis in said cell, reducing expression of p53 in said cell, reducing levels of a cytokine produced in said cell, reducing levels of a cytokine produced in said cell, increasing expression of Bcl-2 in said cell; reducing levels of myeloperoxidase produced in said cell, reducing levels of active NFk beta in said cell, reducing levels of TLR4 in said cell, reducing levels of TNFR-1 in said cell and reducing levels of iNOS in said cell.
14. The method of claim 12 herein said inhibition of expression of endogenous apoptosis-specific eIF-5A in said cell has an effect on the cell selected from the group consisting of suppressing apoptosis in said cell, reducing expression of p53 in said cell, reducing levels of a cytokine produced in said cell, reducing levels of a cytokine produced in said cell, increasing expression of Bcl-2 in said cell; reducing levels of myeloperoxidase produced in said cell, and reducing levels of active NFk beta in said cell, reducing levels of TLR4 in said cell, reducing levels of TNFR-1 in said cell and reducing levels of iNOS in said cell.
15. A method delivering siRNA to lung cells of a mammal in vivo, the method comprising mixing said siRNA with water and delivering to a mammal intranasally.
16. An siRNA of apoptosis-specific eIF-5A wherein said siRNA suppresses endogenous expression of apoptosis-specific eIF-5A in a cell.
17. The siRNA of claim 16, wherein apoptosis-specific eIF-5A is encoded by nucleotide sequence of SEQ ID NO:20.
18. The siRNA of claim 16, wherein apoptosis-specific eIF-5A is encoded by nucleotide sequence of SEQ ID NO:41.
19. The siRNA of claim 16 wherein the siRNA has the sequence set forth in SEQ ID NO:30.
20. The siRNA of claim 16 wherein the siRNA has the sequence set forth in SEQ ID NO:31.
21. The siRNA of claim 16 wherein the siRNA has the sequence set forth in SEQ ID NO:32.
22. The siRNA of claim 16 wherein the siRNA has the sequence set forth in SEQ ID NO:33.
23. A method of inhibiting expression of apoptosis-specific eIF-5A in a cell, the method comprising administering the siRNA of claim 16 to said cell whereby said apoptosis-specific eIF-5A siRNA inhibits expression of endogenous apoptosis-specific eIF-5A in said cell.
24. A method of inhibiting expression of apoptosis-specific eIF-5A in a cell, the method comprising administering the siRNA of claim 19, 20, 21, or 22 to said cell whereby said apoptosis-specific eIF-5A siRNA inhibits expression of endogenous apoptosis-specific eIF-5A in said cell.
25. A method of inhibiting expression of apoptosis-specific eIF-5A in a cell, the method comprising administering the siRNA of claim 16 to said cell whereby said apoptosis-specific eIF-5A siRNA inhibits expression of endogenous apoptosis-specific eIF-5A in said cell.
26. A method of inhibiting expression of apoptosis-specific eIF-5A in a cell, the method comprising administering the siRNA of claim 19, 20, 21, or 22 to said cell whereby said apoptosis-specific eIF-5A siRNA inhibits expression of endogenous apoptosis-specific eIF-5A in said cell.
27. The method of claim 25 wherein said inhibition of expression of endogenous apoptosis-specific eIF-5A in said cell has an effect on the cell selected from the group consisting of suppressing apoptosis in said cell, reducing expression of p53 in said cell, reducing levels of a cytokine produced in said cell, reducing levels of a cytokine produced in said cell, increasing expression of Bcl-2 in said cell; reducing levels of myeloperoxidase produced in said cell, and reducing levels of active NFk beta in said cell, reducing levels of TLR4 in said cell, reducing levels of TNFR-1 in said cell and reducing levels of iNOS in said cell.
28. The method of claim 26 herein said inhibition of expression of endogenous apoptosis-specific eIF-5A in said cell has an effect on the cell selected from the group consisting of suppressing apoptosis in said cell, reducing expression of p53 in said cell, reducing levels of a cytokine produced in said cell, reducing levels of a cytokine produced in said cell, increasing expression of Bcl-2 in said cell; reducing levels of myeloperoxidase produced in said cell, and reducing levels of active NFk beta in said cell, reducing levels of TLR4 in said cell, reducing levels of TNFR-1 in said cell and reducing levels of iNOS in said cell.
US11/134,445 2001-07-23 2005-05-23 Inhibition of apoptosis-specific eIF-5A ("eIF-5A1") with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics Abandoned US20100168047A9 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US11/134,445 US20100168047A9 (en) 2001-07-23 2005-05-23 Inhibition of apoptosis-specific eIF-5A ("eIF-5A1") with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics
US11/184,982 US20060154887A1 (en) 2001-07-23 2005-07-20 Use of apoptosis-specific eIF-5A siRNAs and antisense polynucleotides to inhibit/suppress an inflammatory response
US13/099,171 US8754057B2 (en) 2003-03-05 2011-05-02 Inhibition of apoptosis-specific eIF-5A(eIF-5A1″) with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics
US14/475,389 US20140371299A1 (en) 2004-06-07 2014-09-02 Use of Apoptosis-Specific elF-5A siRNA to Down Regulate Expression of Proinflammatory Cytokines to Treat Sepsis

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US09/909,796 US6867237B1 (en) 2001-07-23 2001-07-23 DNA encoding apoptosis-induced eucaryotic initiation factor-5A and deoxyhypusine synthase and a method for controlling apoptosis in animals and humans
US10/141,647 US7166467B2 (en) 2001-07-23 2002-05-07 Nucleic acids, polypeptides, compositions, and methods for modulating apoptosis
US10/200,148 US7968523B2 (en) 2001-07-23 2002-07-23 Method for inducing apoptosis using apoptosis-specific EIF5-A
US10/277,969 US7217517B2 (en) 2001-07-23 2002-10-23 Nucleic acids, polypeptides, and methods for modulating apoptosis
US45167703P 2003-03-05 2003-03-05
US10/383,614 US7381708B2 (en) 2001-07-23 2003-03-10 Suppression of eIF5A1 expression by the use of antisense oligonucleotides for the prevention of retinal cell death in the glaucomatous eye
US47619403P 2003-06-06 2003-06-06
US50473103P 2003-09-22 2003-09-22
US52824903P 2003-12-10 2003-12-10
US55767104P 2004-03-31 2004-03-31
US86198004A 2004-06-07 2004-06-07
US11/134,445 US20100168047A9 (en) 2001-07-23 2005-05-23 Inhibition of apoptosis-specific eIF-5A ("eIF-5A1") with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
US10/277,969 Continuation-In-Part US7217517B2 (en) 2001-07-23 2002-10-23 Nucleic acids, polypeptides, and methods for modulating apoptosis
US10/383,614 Continuation-In-Part US7381708B2 (en) 2001-07-23 2003-03-10 Suppression of eIF5A1 expression by the use of antisense oligonucleotides for the prevention of retinal cell death in the glaucomatous eye
US86198004A Continuation 2001-07-23 2004-06-07

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US11/184,982 Continuation-In-Part US20060154887A1 (en) 2001-07-23 2005-07-20 Use of apoptosis-specific eIF-5A siRNAs and antisense polynucleotides to inhibit/suppress an inflammatory response
US13/099,171 Continuation US8754057B2 (en) 2003-03-05 2011-05-02 Inhibition of apoptosis-specific eIF-5A(eIF-5A1″) with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics

Publications (2)

Publication Number Publication Date
US20060094677A1 true US20060094677A1 (en) 2006-05-04
US20100168047A9 US20100168047A9 (en) 2010-07-01

Family

ID=32966688

Family Applications (5)

Application Number Title Priority Date Filing Date
US11/134,445 Abandoned US20100168047A9 (en) 2001-07-23 2005-05-23 Inhibition of apoptosis-specific eIF-5A ("eIF-5A1") with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics
US11/287,460 Expired - Fee Related US7662796B2 (en) 2001-07-23 2005-11-28 Use of antisense oligonucleotides or siRNA to suppress expression of eIF-5A1
US12/640,148 Expired - Fee Related US8242256B2 (en) 2001-07-23 2009-12-17 siRNA useful to supress expression of EIF-5A1
US13/099,171 Expired - Fee Related US8754057B2 (en) 2003-03-05 2011-05-02 Inhibition of apoptosis-specific eIF-5A(eIF-5A1″) with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics
US13/572,123 Abandoned US20130066058A1 (en) 2001-07-23 2012-08-10 Use of Antisense Oligonucleotides or siRNA to Suppress Expression of eIF-5A1

Family Applications After (4)

Application Number Title Priority Date Filing Date
US11/287,460 Expired - Fee Related US7662796B2 (en) 2001-07-23 2005-11-28 Use of antisense oligonucleotides or siRNA to suppress expression of eIF-5A1
US12/640,148 Expired - Fee Related US8242256B2 (en) 2001-07-23 2009-12-17 siRNA useful to supress expression of EIF-5A1
US13/099,171 Expired - Fee Related US8754057B2 (en) 2003-03-05 2011-05-02 Inhibition of apoptosis-specific eIF-5A(eIF-5A1″) with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics
US13/572,123 Abandoned US20130066058A1 (en) 2001-07-23 2012-08-10 Use of Antisense Oligonucleotides or siRNA to Suppress Expression of eIF-5A1

Country Status (9)

Country Link
US (5) US20100168047A9 (en)
EP (1) EP1601767B1 (en)
JP (1) JP2006520611A (en)
KR (1) KR20050098954A (en)
AU (1) AU2004217437B2 (en)
CA (1) CA2517974A1 (en)
NZ (1) NZ542665A (en)
TW (1) TW200427695A (en)
WO (1) WO2004078940A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080287381A1 (en) * 2005-01-30 2008-11-20 Sebastian Thaler Injectable Agent for the Targeted Treatment of Retinal Ganglion Cells
US20090118207A1 (en) * 2006-03-20 2009-05-07 Senesco Technologies, Inc. Use of apoptosis-specific eIF-5A siRNA to down regulate expression of proinflammatory cytokines to treat sepsis
WO2010118224A1 (en) * 2009-04-08 2010-10-14 Indiana University Research And Technology Corporation Preventing islet inflammation and dysfunction and maintaining proper glucose levels by controlling elf5a and its hypusination
US20110177089A1 (en) * 2008-05-27 2011-07-21 The University Of Tokyo Apoptosis inducer

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7381708B2 (en) 2001-07-23 2008-06-03 Sensco Technologies, Inc. Suppression of eIF5A1 expression by the use of antisense oligonucleotides for the prevention of retinal cell death in the glaucomatous eye
EP1601767B1 (en) 2003-03-05 2012-04-25 Senesco Technologies, Inc. USE OF siRNA TO SUPPRESS EXPRESSION OF EIF-5A1 IN THE TREATMENT OF GLAUCOMA
ATE555201T1 (en) * 2003-03-05 2012-05-15 Senesco Technologies Inc USE OF SIRNA TO ABOLITATE EXPRESSION OF EIF-5A1 IN THE TREATMENT OF GLAUCOMA
NZ544159A (en) * 2003-06-06 2009-04-30 Senesco Technologies Inc Inhibition of apoptosis-specific eIF-5A ("eIF-5A1") with antisense oligonucleotides and siRNAs as anti-inflammatory therapeutics
US20140371299A1 (en) * 2004-06-07 2014-12-18 Senesco Technologies, Inc. Use of Apoptosis-Specific elF-5A siRNA to Down Regulate Expression of Proinflammatory Cytokines to Treat Sepsis
JP2008507278A (en) * 2004-07-20 2008-03-13 セネスコ テクノロジーズ,インコーポレイティド Use of apoptosis-specific eIF-5A siRNA and antisense polynucleotides to inhibit / suppress inflammatory responses
TW200634154A (en) * 2004-12-03 2006-10-01 Senesco Technologies Inc Apoptosis-specific EIF-5A and polynucleotides encoding same
CA2588330A1 (en) * 2004-12-03 2006-06-08 Senesco Technologies, Inc. Methods of increasing seed yield with a growth eif-5a
US7615554B2 (en) 2005-01-25 2009-11-10 Prolexys Pharmaceuticals, Inc. Erastin and erastin binding proteins, and uses thereof
US8105611B2 (en) * 2005-06-17 2012-01-31 Allergan, Inc. Treatment of autoimmune disorder with a neurotoxin
WO2007070824A2 (en) * 2005-12-13 2007-06-21 Senesco Technologies, Inc. Use of eif-5a to kill multiple myeloma cells
JP5543774B2 (en) * 2006-03-20 2014-07-09 セネスコ テクノロジーズ,インコーポレイティド A novel method to prevent islet cells from being apoptotic during the donor harvest process
US20070238691A1 (en) * 2006-03-29 2007-10-11 Senesco Technologies, Inc. Inhibition of HIV replication and expression of p24 with eIF-5A
ATE508191T1 (en) * 2006-11-01 2011-05-15 Medical Res And Infrastructure Fund Of The Tel Aviv Sourasky Medical Ct ADIPOCYTE-SPECIFIC CONSTRUCTS AND METHODS FOR INHIBITING THE EXPRESSION OF PLATELET TYPE 12-LIPOXYGENASE
WO2008065429A1 (en) * 2006-11-30 2008-06-05 Ucl Business Plc Method for delivering gene therapy vectors to the optic nerve head
CN101126098B (en) * 2007-07-05 2012-07-11 深圳市第二人民医院 Small disturbance RNA molecule expression carrier for differentially inhibiting GFAP protein expression and its construction method and use thereof
CN102282259A (en) 2008-03-07 2011-12-14 森尼斯科技术公司 Use of SIRNA to achieve down regulation of an endogenous gene in combination with the use of a sense construct to achieve expression of a desired polynucleotide
US8946239B2 (en) 2008-07-10 2015-02-03 Duquesne University Of The Holy Spirit Substituted pyrrolo, -furano, and cyclopentylpyrimidines having antimitotic and/or antitumor activity and methods of use thereof
US8445638B2 (en) 2008-09-03 2013-05-21 Senesco Technologies, Inc. Use of a truncated eIF-5A1 polynucleotide to induce apoptosis in cancer cells
CA2738957A1 (en) 2008-10-22 2010-04-29 Quark Pharmaceuticals, Inc. Methods for treating eye disorders
NZ733634A (en) 2009-12-01 2022-10-28 Translate Bio Inc Delivery of mrna for the augmentation of proteins and enzymes in human genetic diseases
RS59037B1 (en) 2011-06-08 2019-08-30 Translate Bio Inc Lipid nanoparticle compositions and methods for mrna delivery
EP3536787A1 (en) 2012-06-08 2019-09-11 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
DK2970955T3 (en) 2013-03-14 2019-02-11 Translate Bio Inc METHODS FOR CLEANING MESSENGER RNA
KR20210122917A (en) 2013-03-14 2021-10-12 샤이어 휴먼 지네틱 테라피즈 인크. Cftr mrna compositions and related methods and uses
SG11201507496UA (en) 2013-04-17 2015-11-27 Pfizer N-piperidin-3-ylbenzamide derivatives for treating cardiovascular diseases
CN103421791B (en) * 2013-06-24 2015-04-15 广西医科大学 SiRNA of inhibiting expression of TLR2 of human cytomegalovirus and application thereof
AU2014340083B2 (en) 2013-10-22 2019-08-15 Translate Bio, Inc. mRNA therapy for phenylketonuria
CN106413811A (en) 2013-10-22 2017-02-15 夏尔人类遗传性治疗公司 Mrna therapy for argininosuccinate synthetase deficiency
EP3134506B1 (en) 2014-04-25 2019-08-07 Translate Bio, Inc. Methods for purification of messenger rna
EA201991747A1 (en) 2017-02-27 2020-06-04 Транслейт Био, Инк. NEW CODON-OPTIMIZED CFTR mRNA
WO2018213476A1 (en) 2017-05-16 2018-11-22 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mrna encoding cftr
WO2019100005A1 (en) * 2017-11-18 2019-05-23 De Magalhaes Nzola Product and process for employing gc7 (n1-guanyl-1,7-diaminoheptane) based antigen binding conjugates in cancer therapy
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
CN114231551B (en) * 2021-12-24 2023-09-29 云南大学 Application of protein in promoting apoptosis of insect lymphocytes and/or preventing and controlling pests

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6274630B1 (en) * 1996-09-13 2001-08-14 University Of Florida Research Foundation, Inc. Method of inhibiting biosynthesis of EIf5A
US20030144238A1 (en) * 2001-07-23 2003-07-31 Thompson John E. Nucleic acids, polypeptides, and methods for modulating apoptosis
US20030225022A1 (en) * 2001-07-23 2003-12-04 Catherine Taylor Suppression of eIF5A1 expression to prevent retinal ganglion cell death in the glaucomatous eye
US20070031844A1 (en) * 2002-11-14 2007-02-08 Anastasia Khvorova Functional and hyperfunctional siRNA

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR19980702160A (en) 1995-02-13 1998-07-15 하우스 한스 루돌프. 나톨리 안토니오 Eukaryotic Initiation Factor 5A (IIF-5A) Mutants
EP1002079A2 (en) 1997-06-30 2000-05-24 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Novel inhibitor of cellular proliferation
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US7358418B2 (en) 1999-07-06 2008-04-15 Senesco Technologies, Inc. Isoforms of eIF-5A: senescence-induced eLF5A; wounding-induced eIF-4A; growth eIF-5A; and DHS
CA2376065A1 (en) 1999-08-06 2001-02-15 Pharmacia & Upjohn Company Crystallization and structure determination of staphylococcus aureus elongation factor p
DE60140676D1 (en) * 2000-03-30 2010-01-14 Massachusetts Inst Technology RNA INTERFERENCE MEDIATORS WHICH ARE RNA SEQUENCE SPECIFIC
DE60130583T3 (en) * 2000-12-01 2018-03-22 Europäisches Laboratorium für Molekularbiologie SMALL RNA MOLECULES TRANSFERRING RNA INTERFERENCE
JP2003037457A (en) * 2001-07-23 2003-02-07 Niigata Seimitsu Kk Amplifier circuit
US7968523B2 (en) * 2001-07-23 2011-06-28 Senesco Technologies, Inc. Method for inducing apoptosis using apoptosis-specific EIF5-A
US7166467B2 (en) * 2001-07-23 2007-01-23 Senesco Technologies, Inc. Nucleic acids, polypeptides, compositions, and methods for modulating apoptosis
WO2003016572A1 (en) * 2001-08-17 2003-02-27 Eli Lilly And Company Oligonucleotide therapeutics for treating hepatitis c virus infections
US20030166282A1 (en) * 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
EP1601767B1 (en) 2003-03-05 2012-04-25 Senesco Technologies, Inc. USE OF siRNA TO SUPPRESS EXPRESSION OF EIF-5A1 IN THE TREATMENT OF GLAUCOMA
NZ544159A (en) 2003-06-06 2009-04-30 Senesco Technologies Inc Inhibition of apoptosis-specific eIF-5A ("eIF-5A1") with antisense oligonucleotides and siRNAs as anti-inflammatory therapeutics

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6274630B1 (en) * 1996-09-13 2001-08-14 University Of Florida Research Foundation, Inc. Method of inhibiting biosynthesis of EIf5A
US20030144238A1 (en) * 2001-07-23 2003-07-31 Thompson John E. Nucleic acids, polypeptides, and methods for modulating apoptosis
US20030225022A1 (en) * 2001-07-23 2003-12-04 Catherine Taylor Suppression of eIF5A1 expression to prevent retinal ganglion cell death in the glaucomatous eye
US7217517B2 (en) * 2001-07-23 2007-05-15 Senesco Technologies, Inc. Nucleic acids, polypeptides, and methods for modulating apoptosis
US7381708B2 (en) * 2001-07-23 2008-06-03 Sensco Technologies, Inc. Suppression of eIF5A1 expression by the use of antisense oligonucleotides for the prevention of retinal cell death in the glaucomatous eye
US20070031844A1 (en) * 2002-11-14 2007-02-08 Anastasia Khvorova Functional and hyperfunctional siRNA

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080287381A1 (en) * 2005-01-30 2008-11-20 Sebastian Thaler Injectable Agent for the Targeted Treatment of Retinal Ganglion Cells
US20090118207A1 (en) * 2006-03-20 2009-05-07 Senesco Technologies, Inc. Use of apoptosis-specific eIF-5A siRNA to down regulate expression of proinflammatory cytokines to treat sepsis
US8193158B2 (en) * 2006-03-20 2012-06-05 Senesco Technologies, Inc. Use of apoptosis-specific eIF-5A siRNA to down regulate expression of proinflammatory cytokines to treat sepsis
US20110177089A1 (en) * 2008-05-27 2011-07-21 The University Of Tokyo Apoptosis inducer
US8492521B2 (en) * 2008-05-27 2013-07-23 Yoshinori Seko Oxidative stress responsive apoptosis inducing protein eIF5A
US9321819B2 (en) 2008-05-27 2016-04-26 Yoshinori Seko Oxidative stress responsive apoptosis inducing protein eIF5A
WO2010118224A1 (en) * 2009-04-08 2010-10-14 Indiana University Research And Technology Corporation Preventing islet inflammation and dysfunction and maintaining proper glucose levels by controlling elf5a and its hypusination

Also Published As

Publication number Publication date
WO2004078940A3 (en) 2004-12-16
KR20050098954A (en) 2005-10-12
US7662796B2 (en) 2010-02-16
US8754057B2 (en) 2014-06-17
CA2517974A1 (en) 2004-09-16
WO2004078940A2 (en) 2004-09-16
US20130066058A1 (en) 2013-03-14
US20110098460A1 (en) 2011-04-28
US8242256B2 (en) 2012-08-14
JP2006520611A (en) 2006-09-14
US20120129910A1 (en) 2012-05-24
NZ542665A (en) 2008-05-30
TW200427695A (en) 2004-12-16
US20060178330A1 (en) 2006-08-10
EP1601767A2 (en) 2005-12-07
US20100168047A9 (en) 2010-07-01
AU2004217437A1 (en) 2004-09-16
EP1601767B1 (en) 2012-04-25
WO2004078940A8 (en) 2005-10-27
AU2004217437B2 (en) 2009-11-19

Similar Documents

Publication Publication Date Title
US8754057B2 (en) Inhibition of apoptosis-specific eIF-5A(eIF-5A1″) with antisense oligonucleotides and siRNA as anti-inflammatory therapeutics
AU2004258070B2 (en) Inhibition of apoptosis-specific elF-5A ("elF-5A1") with antisense oligonucleotides and siRNAs as anti-inflammatory therapeutics
EP1959010A2 (en) Apoptosis-specific eif-5A and polynucleotides encoding same
US20060287265A1 (en) Apoptosis-specific eIF-5A and polynucleotides encoding same
AU2005269647B2 (en) Use of apoptosis-specific eIF-5A siRNAs and antisense polynucleotides to inhibit/suppress an inflammatory response
US7381708B2 (en) Suppression of eIF5A1 expression by the use of antisense oligonucleotides for the prevention of retinal cell death in the glaucomatous eye
JP5566250B2 (en) Use of antisense oligonucleotides or siRNAs to suppress the expression of eIF-5A1
KR20070018146A (en) Use of apoptosis-specific eIF-5A siRNAs and antisense polynucleotides to inhibit/suppress an inflammatory response

Legal Events

Date Code Title Description
AS Assignment

Owner name: SENESCO TECHNOLOGIES, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:THOMPSON, JOHN E.;GALTON, BRUCE C.;TAYLOR, CATHERINE;AND OTHERS;REEL/FRAME:017591/0872;SIGNING DATES FROM 20050804 TO 20050810

Owner name: SENESCO TECHNOLOGIES, INC.,NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:THOMPSON, JOHN E.;GALTON, BRUCE C.;TAYLOR, CATHERINE;AND OTHERS;SIGNING DATES FROM 20050804 TO 20050810;REEL/FRAME:017591/0872

Owner name: SENESCO TECHNOLOGIES, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:THOMPSON, JOHN E.;GALTON, BRUCE C.;TAYLOR, CATHERINE;AND OTHERS;SIGNING DATES FROM 20050804 TO 20050810;REEL/FRAME:017591/0872

AS Assignment

Owner name: YA GLOBAL INVESTMENTS, L.P., NEW JERSEY

Free format text: SECURITY AGREEMENT;ASSIGNORS:SENESCO TECHNOLOGIES, INC.;SENESCO, INC.;REEL/FRAME:020468/0001

Effective date: 20070921

Owner name: YA GLOBAL INVESTMENTS, L.P.,NEW JERSEY

Free format text: SECURITY AGREEMENT;ASSIGNORS:SENESCO TECHNOLOGIES, INC.;SENESCO, INC.;REEL/FRAME:020468/0001

Effective date: 20070921

AS Assignment

Owner name: SENESCO TECHNOLOGIES, INC.,NEW JERSEY

Free format text: RELEASE OF SECURITY INTEREST IN INTELLECTUAL PROPERTY;ASSIGNOR:YA GLOBAL INVESTMENTS, L.P.;REEL/FRAME:024128/0676

Effective date: 20100324

Owner name: SENESCO, INC.,NEW MEXICO

Free format text: RELEASE OF SECURITY INTEREST IN INTELLECTUAL PROPERTY;ASSIGNOR:YA GLOBAL INVESTMENTS, L.P.;REEL/FRAME:024128/0676

Effective date: 20100324

Owner name: SENESCO TECHNOLOGIES, INC., NEW JERSEY

Free format text: RELEASE OF SECURITY INTEREST IN INTELLECTUAL PROPERTY;ASSIGNOR:YA GLOBAL INVESTMENTS, L.P.;REEL/FRAME:024128/0676

Effective date: 20100324

Owner name: SENESCO, INC., NEW MEXICO

Free format text: RELEASE OF SECURITY INTEREST IN INTELLECTUAL PROPERTY;ASSIGNOR:YA GLOBAL INVESTMENTS, L.P.;REEL/FRAME:024128/0676

Effective date: 20100324

STCB Information on status: application discontinuation

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