GB2526767A - Combined inhibition of caspase-2 and caspase-6 in neuroprotection and axon regeneration - Google Patents

Combined inhibition of caspase-2 and caspase-6 in neuroprotection and axon regeneration Download PDF

Info

Publication number
GB2526767A
GB2526767A GB1404289.9A GB201404289A GB2526767A GB 2526767 A GB2526767 A GB 2526767A GB 201404289 A GB201404289 A GB 201404289A GB 2526767 A GB2526767 A GB 2526767A
Authority
GB
United Kingdom
Prior art keywords
caspase
inhibitor
use according
injury
retinal
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.)
Withdrawn
Application number
GB1404289.9A
Other versions
GB201404289D0 (en
Inventor
Zubair Ahmed
Ann Logan
Martin Berry
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.)
University of Birmingham
Original Assignee
University of Birmingham
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Birmingham filed Critical University of Birmingham
Priority to GB1404289.9A priority Critical patent/GB2526767A/en
Publication of GB201404289D0 publication Critical patent/GB201404289D0/en
Publication of GB2526767A publication Critical patent/GB2526767A/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/185Nerve growth factor [NGF]; Brain derived neurotrophic factor [BDNF]; Ciliary neurotrophic factor [CNTF]; Glial derived neurotrophic factor [GDNF]; Neurotrophins, e.g. NT-3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4873Cysteine endopeptidases (3.4.22), e.g. stem bromelain, papain, ficin, cathepsin H
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/317Chemical structure of the backbone with an inverted bond, e.g. a cap structure
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/344Position-specific modifications, e.g. on every purine, at the 3'-end
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/22Cysteine endopeptidases (3.4.22)
    • C12Y304/22055Caspase-2 (3.4.22.55)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/22Cysteine endopeptidases (3.4.22)
    • C12Y304/22059Caspase-6 (3.4.22.59)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Neurosurgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Psychology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The use of agents for the inhibition of caspase-2 and caspase-6 in the protection of central and peripheral neurons is disclosed. In particular, the invention comprises an inhibitor of caspase-2 for use in the treatment of a disease, a disorder or an injury of the nervous system by combined, sequential or separate administration with an inhibitor of caspase-6. Further provided is a composition comprising an inhibitor of caspase-2 and an inhibitor of caspase-6. The diseases may include ocular disorders or injuries, including ischemic optic neuropathy (ION) or other optical neuropathies.

Description

Intellectual Property Office Application No. GB1404259.9 RTTVT Date:30 September 2015 The following terms are registered trade marks and should be read as such wherever they occur in this document: Superfrost (page 17) Axioplan (page 17, 18, 19) Axiovision (page 17, 19, 20-protected) Image Pro (page 17, 20) Tween (page 1 8) Alexa Fluor (page 18) Texas Red (page 18) Triton (page 19) Axiocam (page 17 & 19-protected) Intellectual Property Office is an operating name of the Patent Office www.gov.uk/ipo COMBINED INHIBITION OF CASPASE-2 AND CASPASE-6 IN
NEUROPROTECTION AND AXON REGENERATION
FIELD AND BACKGROUND OF THE INVENTION 5
The invention, in some embodiments, relates to the use of agents for inhibition of caspase-2 and caspase-6 in protection of central and peripheral neurons, such as retinal ganglion cells (RGC) and dorsal root ganglion neurons (DRGN), from apoptosis and promotion of axon regeneration.
Unlike peripheral nervous system axons, central nervous system (CNS) axons fail to 10 regenerate after injury. Many growth-limiting factors have been implicated, including apoptosis of axotomised neurons, intrinsic blocks to growth, limiting supplies of neurotrophic factors and the presence of an axon growth inhibitory environment in the CNS neuropil.
Optic nerve (ON) injury induces progressive RGC death (Villegas-Perez ci at, 1993, Berkelaar ci at., 1994, Kermer ci at, 1998), so that 70-75% of RGC are lost within 7 days 15 (Berry c/at, t996, Berry cia!., 1999, Agudo eta)., 2008, Abmed c/at, 2Ott, Vigneswaraci at, 2012) and 80-90% by 28 days, primarily as a result of apoptosis (Garcia-Valenzuela ci at, t994, Rabacchi ci at, 1994, Isenmann ci at, 1997).
Caspases are a family of cysteine-dependent proteases well known for their orchestration of apoptosis, although they also have non-apoptotic roles. They are expressed as pro-caspases 20 and activated by either proximity induced dimerization (initiator caspases) or proteolytic cleavage (effector caspases) (Pop and Salvesen, 2009). This enables active caspase enzymes to cleave regulatory proteins that play roles in DNA replication (Casciola-Rosen c/at, 1994, Song eta!.., 1996), DNA repair (Lazebnik ci at, 1994), cell survival signalling (Leist ci at, 1997), cytoskeletal reorganisation and cellular disassembly (Porter ci a!., 1997, Bokoch, 25 1998).
Injured RGC exclusively express and cleave caspase-2 (Ahmed ci at, 2011, Vigneswara ci at, 2012) and suppression of caspase-2, by either a chemically stabilised caspase-2 siRNA (siCASP2) or a pharmacological inhibitor, significantly protects RGC from death for at least 14 days after injury (Ahmed c/at, 2011, Vigneswara c/at, 2012). Despite significant RGC 30 neuroprotection by both of these pharmacological strategies, RGC axon regeneration is unaffected, suggesting that different signalling pathways regulate neuron survival and axon regeneration. The present inventors have shown previously that the cues for post-injury ROC survival and axon regeneration, which can be mediated by inflammation, are different, since both retinal and ON inflammation promotes RGC survival but only retinal inflammation is RUC axogenic (Ahmed et aL, 2010). RGC survival and axon regeneration require the activation of both survival and growth signalling pathways. 5 SUTVHVIAB.Y OF THE INVENTION Some embodiments of the invention relate to use of agents for combined inhibition of caspase-2 and caspase-6 for use in the treatment of a disease, a disorder or an injury of the nervous system by combined, sequential or separate administration with an inhibitor of caspase-6, Aspects and embodiments of the invention are described in the specification herein below and in the appended claims.
Some embodiments of the invention relate to the use of an inhibitor of caspase-2 and an Jo inhibitor of caspase-6 in therapy.
According to an aspect of some embodiments of the invention, there is provided an inhibitor of caspase-2 for use in the treatment of a disease, a disorder or an injury of the nervous system by combined, sequential or separate administration with an inhibitor of caspase-6.
According to an aspect of some embodiments of the invention, there is provided a method of treating a disease, a disorder or an injury of the nervous system by combined, sequential or separate administration of an inhibitor of caspase-2 and an inhibitor of caspase-6, In some embodiments, the nervous system is selected from the group consisting of the central nervous system (CNS) and the peripheral nervous system (PNS).
In some embodiments, the central nervous system (CNS) comprises the visual system and the audlo-vestibular system.
In some embodiments, the disease, disorder or injury of the CNS is selected from the group consisting of an ocular disease; an ocular disorder or an ocular injury; a spinal cord disease, a spinal cord disorder or a spinal cord injury; a brain disease, a brain disorder or a brain injury; and a disease, disorder or injury of the audio-vestibular system, or a combination thereof In some embodiments, the disease, disorder or injury of the CNS is selected from the group consisting of a neurodegenerative disease (such as an acute neurodegenerative disease or a chronic neurodegenerative disease) and a neurological disorder, or a combination thereof In some embodiments, the disease, disorder or injury of the CNS is selected from the group consisting of inflammation, neurotoxicity, oxidative stress, and traumatic damage due to a tumor growth, or a combination thereof In some embodiments, the disease, disorder or injury of the CNS is an ocular disease, an ocular disorder or an ocubr injury.
In some embodiments, the ocular disease, ocular disorder or ocular injury is selected from the group consisting of neurodegeneration, inflammation, and oxidative stress, or a combination thereof In some embodiments, the ocular disease, ocular disorder or ocular injury is selected from the group consisting of glaucoma (including open angle glaucoma and angle closure glaucoma), and diabetic retinopathy (DR) or a combination thereof In some embodiments, the ocular disease, ocular disorder or ocular injury comprises an optic neuropathy selected from the group consisting of ischemic optic neuropathy (ION) (such as non-arteritic ischemic optic neuropathy (NAION), hereditary optic neuropathy (such asLeber's hereditary optic neuropathy (LHON)), metabolic optic neuropathy, neuropathy due to a toxic agent, neuropathy caused by adverse dmg reactions and neuropathy caused by vitamin deficiency, or a combination thereof In some embodiments, the ocular disease, ocular disorder or ocular injury is selected from the group consisting of optic neuritis, retinal artery occlusion, central retinal vein occlusion, brunch retinal vein occlusion and optic nerve injury, or a combination thereof In some embodiments, the ocular disease, ocular disorder or ocular injury is selected from the group consisting of ischemic injury, ischemia-reperfusion injury, mechanical injury, injury or intermption of nerve fibers, physically damaged nerve, neurite damage, and a condition associated with lack of retrograde supply of neurotrophic factor, or a combination thereof In some embodiments, the treatment provides neuroprotection, In some embodiments, the treatment provides axon regeneration of neuronal cells, such as axon regeneration of retinal neuronal cells or dorsal root ganglion neurons, In some embodiments, the treatment provides both neuroprotection and axon regeneration of neuronal cells.
In some embodiments, the neuronal cells are cells of the peripheral nervous system (PNS), In some embodiments, the neuronal cells are celis of the central nervous system (CNS).
In some embodiments, the neuroprotection comprises ocular neuroprotection, such as audio-vestibular neuroprotection.
In some embodiments, the neuroprotection is selected from the group consisting of protection of neural cells from apoptosis, promoting survival of neural cells, increasing the number of neural cell neuritis, increasing neurite cell outgrowth, promoting retinal gliosis, promoting regeneration of neural cells and increasing or stimulation of neurotrophic factors in the nervous system, or a combination thereof In some embodiments, the neural cell comprises a ganglion cell, such as a ganglion cell selected from the group consisting of a retinal ganglion cell, a spiral ganglion cell, a vestibular ganglion cell, a dorsal root ganglion cell and a peripheral ganglion cell, or a combination thereof In some embodiments, the neural cell comprises a retinal neural cell, such as a photoreceptor cell, a bipolar cell, a ganglion cell, a horizontal cell and an amacrine cell, or a combination thereof In some embodiments, the neural cell comprises a dorsal root ganglion cell.
In some embodiments, retinal neural cell comprises a retinal ganglion cell (RGC), In some embodiments, neural cell comprises a dorsal root ganglion cell (DRGC).
In some embodiments, the neuroprotection comprises protecting neurons from death. In some such embodiments, death of the neuron is associated with one or more of a disease or disorder, a surgery, ischemia, ischemialreperfusion, physical/mechanical trauma, a chemical agent, an infectious agent, an immunologic reaction and a nutritional imbalance.
In some embodiments, the inhibitor of caspase-2 and the inhibitor of caspase-6 are independently configured for contacting the neuron.
According to an aspect of some embodiments disclosed herein, there is provided an inhibitor of caspase-2 for use in providing neuroprotection and axon regeneration of neuronal cells by combined, sequential or separate administration with an inhibitor of caspase-6.
According to an aspect of some embodiments of the invention, there is provided a composition comprising an inhibitor of caspase-2 and an inhibitor of caspase-6.
In some embodiments, the inhibitor of caspase-2 and the inhibitor of caspase-6 are independently selected from the group consisting of a small organic molecule, a protein, an antibody or fragment thereof, a peptide, a polypeptide, a peptidomimetic and a nucleic acid molecule; or a salt or prodrug thereof; or a combination thereof In some embodiments, at least one of the inhibitor of caspase-2 and the inhibitor of caspase-6 is a nucleic acid molecule independently selected from the group consisting of a single stranded antisense nucleic acid (ssNA), a double-stranded NA (dsNA), a small interfering NA (siNA), a short hairpin NA (shNA), a micro RNA (miRNA), an aptamer, and a ribozyme, or a salt or prodmg thereof, or a combination thereof In some embodiments, each of the ssNA or the dsNA independently comprises one or more of a modified nucleotide, an unmodified nucleotide, a nucleotide analogue and an unconventional moiety, such as an RNA, a DNA, a TNA or an ANA. In some embodiments, the inhibitor of caspase-2 is a dsRNA, In some embodiments, at least one of the inhibitor of caspase-2 and the inhibitor of caspase-6 is a double-stranded NA (dsNA) independently selected from an unmodified double-stranded NA (dsNA) or a chemically modified double-stranded NA (dsNA); or a salt or prodrug thereof.
The nucleotides can be selected from naturally occurring or synthetic modified bases.
Naturally occurring bases include adenine, guanine, cytosine, thymine and uracil, Modified bases of nucleotides include inosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl, 2-5 propyl and other alkyl adenines, 5-halo uracil, 5-halo cytosine, 6-aza cytosine and 6-aza thymine, pseudo uracil, 4-thiouracil, 8-halo adenine, 8-aminoadenine, 8-thiol adenine, 8- thiolalkyl adenines, 8-hydroxyl adenine and other 8-substituted adenines, 8-halo guanines, 8-amino guanine, 8-thiol guanine, 8-thioalkyl guanines, 8-hydroxyl guanine and other substituted guanines, other aza and deaza adenines, other aza and deaza guanines, 5-10 trifluoromethyl uracil and 5-trifluoro cytosine, Molecules comprising one or more abasic moiety (unconventional or pseudonucleotide) are encompassed by the present invention, as well as molecules comprising alternating RNA and DNA nucleotides.
In addition, analogues of polynucleotides can be prepared wherein the structure of one or more nucleotide is thndamentally altered and better suited as therapeutic or experimental 15 reagents. An example of a nucleotide analog is a peptide nucleic acid (PNA) wherein the deoxyribose (or ribose) phosphate backbone in DNA (or RNA) is replaced with a polyamide backbone which is similar to that found in peptides. PNA analogs have been shown to be resistant to enzymatic degradation and to have extended lives in vivo and in vitro.
Possible modifications to the sugar residue are manifold and include 2'-O alkyl, locked 5 nucleic acid (LNA), glycol nucleic acid (GNA), threose nucleic acid (TNA), arabinoside, altritol (ANA) and other, 6-membered sugars including morpholinos, and cyclohexinyls.
Further, said molecules may additionally contain modifications on the sugar, such as 2' alkyl, 2' fluoro, 2'O allyl, 2'amine and 2'aikoxy. Additional sugar modifications are discussed herein. 10 In some embodiments, the inhibitor of caspase-2 comprises an antisense strand sequence 5' AGGAGIJUCCACAL11JCUGGC 3' and a sense strand sequence 5' GCCAGAAUGUGGAACUCCU 3', In some embodiments, the antisense strand of the inhibitor of caspase-2 comprises 2'-O-methyl sugar modified ribonucleotides in positions (5>3') 2, 4, 6, 8, 11, t3, tS, 17 and 19; and wherein the sense strand of the inhibitor of caspase-2 comprises an L-DNA (L-deoxycytidine) nucleotide in position 18 and an inverted deoxyabasic moiety covalently attached at the 5' terminal of the strand, 15 In some embodiments, the inhibitor of caspase-6 comprises caspase-6 dominant negative (C6DN). In some embodiments, C6DN is coupled to a peptide, for example a cell penetrating peptide such as Penetratin-1 (Peni).
The inhibitor of caspase-2 and the inhibitor of caspase-6 may be administered by any of the conventional routes of administration, The chemically modified dsRNA compounds are 20 administered orally, subcutmeously or parenterally including intravenous, intraarterial, intramuscular, intraperitoneally, intraocular, intracoronary, transtympani c, transcomeal, transepithelial, transmembrane and intranasal administration as well as intrathecal and inifision techniques, Implants of the compounds are also useful, In some embodiments, the inhibitor of caspase-2 is for use by concurrent administration with 25 the inhibitor of caspase-6, In some embodiments, the inhibitor of caspase-2 and the inhibitor of caspase-6 are administered in a single dosage form, In some embodiments, the inhibitor of caspase-2 and the inhibitor of caspase-6 are administered in separate dosage forms.
In some embodiments, the inhibitor of caspase-2 is for sequential administration with the inhibitor of caspase-6, In some such embodiments, the inhibitor of caspase-2 is administered prior to administration of the inhibitor of caspase-6. In other such embodiments, the the 5 inhibitor of caspase-2 is administered subsequent to administration of the inhibitor of caspase-6, In some embodiments, the inhibitor of caspase-2 and the inhibitor of caspase-6 comprise different portions of a single molecule.
In some embodiments, there is provided the use of the inhibitor of caspase-2 and the inhibitor 10 of caspase-6 as described herein, for combined, sequential or separate administration with a neurotrophic factor(s), for example but not exclusively, a neurotrophic factor selected from the group consisting of ciliary neurotrophic factor (CNTF), brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3 (NT-3) and fibroblast growth factor-2 (FGF-2), or any combination thereof 15 Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. In case of conflict, the specification, including definitions, takes precedence.
As used herein, the terms "comprising", "including", "having" and grammatical variants thereof are to be taken as specifying the stated features, integers, steps or components but do 20 not preclude the addition of one or more additional features, integers, steps, components or groups thereof These terms encompass the terms "consisting of' and "consisting essentially of,'.
As used herein, the indefinite articles "a" and "an" mean "at least one" or "one or more" unless the context clearly dictates otherwise. 25 As used herein the term "inhibitor" refers to a compound, which is capable of reducing (partially or fully) the expression of a gene or the activity of the product of such gene to an extent sufficient to achieve a desired biological or physiological effect.
As used herein a "therapeutic composition" refers to a preparation of one or more of the active ingredients with other components such as pharmaceutically-acceptable carriers and 31) excipients. The purpose of a therapeutic composition is to facilitate administration of an active ingredient to a subject.
The term "pharmaceutically acceptable carrier" refers to a carrier or a diluent that does not cause significant irritation to a subject and does not substantially abrogate the activity and properties of the administered active ingredients. An adjuvant is induded under these 5 phrases. The term "excipient' refers to an inert substance added to a therapeutic composition to further facilitate administration of an active ingredient.
The term "unconventional moiety" as used herein refers to abasic ribose moiety, an abasic deoxyribose moiety, a deoxyribonucleotide, a modified deoxyribonucleotide, a mirror nucleotide, a non-base pairing nucleotide analog and a nucleotide joined to an adjacent Jo nucleotide by a 2'-S' internucleotide phosphate bond; bridged nucleic acids including LNA and ethylene bridged nucleic acids.
In the context of the present invention, a "miror" nucleotide (also referred to as a spieglemer), is a nucleotide analog with reverse chirality to the naturally occurring or commonly employed nucleotide, i.e., a mirror image of the naturally occurring or commonly 15 employed nucleotide, The mirror nucleotide is a ribonucleotide (L-RNA) or a deoxyribonucleotide (L-DNA) and may further comprise at least one sugar or base modification and/or a backbone modification, such as a phosphorothioate or phosphonate moiety. US Patent No. 6,602,858 discloses nucleic acid catalysts comprising at least one L-nucleotide substitution, Mirror nucleotide includes for example L-DNA (L-20 deoxyriboadenosine-3'-phosphate (mirror dA); L-deoxyribocytidine-3'-phosphate (mirror dC); L-deoxyriboguanosine-3 -phosphate (minor dG); L-deoxyribothymidine-3 -phosphate (mirror dT) and L-RNA (L-riboadenosine-3 -phosphate (mirror rA); L-ribocytidine-3 -phosphate (mirror rC); L-riboguanosine-3 -phosphate (mirror rG); L-ribouraci 1-3 -phosphate (mirror dU). 25 Therapeutic compositions used in implementing the teachings herein may be formulated using techniques with which one of average skill in the art is familiar in a conventional manner using one or more pharmaceutically-acceptable carriers comprising excipients and adjuvants, which facilitate processing of the active ingredients into a therapeutic composition and generally includes mixing an amount of the active ingredients with the other components. 30 Suitable techniques are described in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference. For a example, therapeutic compositions useful in implementing the teachings herein may be manufactured by one or more processes that are well known in the art, e.g., mixing, blending, homogenizing, dissolving, granulating, emulsifying, encapsulating, entrapping and lyophilizing processes.
Therapeutic compositions suitable for implementing the teachings herein include 5 compositions comprising active ingredients in an amount effective to achieve the intended purpose (a therapeutically effective amount). Determination of a therapeutically effective amount is well within the capability of those skilled in the art, for example, is initially estimated from animal models.
BRIEF DESCRIPTION OF THE FIGURES
Some embodiments of the invention are described herein with reference to the accompanying figures. The description, together with the figures, makes apparent to a person having ordinary skill in the art how some embodiments of the invention may be practiced.
In the Figures: 15 FIGs. IA-IF show dose-response relationship for Penl-C6DN. Optic nerves were crushed and the retinae treated with a range of doses of Penl-C6DN from O-7RM C6DN at day 0, 7 and 14 after ONC. At 19d after ONC (FluoroGold) FG was injected into the proximal ON stump and allowed to retrogradely fill surviving RGC. 2 days later, animals were killed, retinae were harvested, wholemounts were made and the number of FG labelled RGC was 20 quantified by image analysis. FIGs. lA-IL are representative photomicrographs of FluoroGold (FG) labeled RGC. FIG. IF is a bar graph showing the dose-response relationship for Pen-C6DN concentration and RGC survival, showing increased RGC survival with increasing concentrations of Penl-C6DN up to 5pM. Scale bar = 50I.tm;*P<0.05, ***p<o 25 FIGs. 2A-2D show the effects of 51.tM Penl-C6DN alone or in combination with siCASP2 on RGC survival, Optimal dose of Penl-C6DN was intravitreally delivered either alone or in combination with siCASP2 immediately after ONC and at 7 and 14 days after ONC. At 21 days, animals were killed and retinae were harvested either for western blot analysis or to count the number of FG-labelled RGC in retinal wholemounts. Western blot analysis (FIG. 30 2A) and subsequent densitometry (FIG. 2B) show that suppression of caspase-6 using 5plVl PenI-C6DN prevents cleavage of Lamin NC while siCASP2 suppresses the levels of caspase-2 in treated eyes. FluoroGold labelling of RGC (FIGs. 2C and 2D0 demonstrate that Penl-CoDN protects nearly 50% of RGC from apoptosis while intravitreal delivery of either Penl+siCASP2 or Penl-C6DN+siCASP2 promoted >c5% of RGC survival. GAPDH was used as a protein loading control. ***P<0 0001 Scale bar in C = 50 jim.
FIGs. 3A-3G show the effects of intravitreal delivery of optimized Penl-C6DN alone or in 5 combination on promotion of RGC axon regeneration. After ONC, optimised Penl-CÔDN either alone or in combination with siCASP2 was intravitreally injected at 0, 7 and 14 days.
Animals were killed at day 21, optic nerves dissected out and processed for immunohistochemistry. FIGs. 3A-3D show GAP-43 stained images of regenerating RGC axons after treatment with Peni alone (3A), Penl-C6DN (3B), Peni+siCASP (3C) and Pen!-10 C6DN+siCASP2 (3D). Inset shows high power magnification of boxed region in 3D. FIG. 3E is a bar graph showing quantification of RGC axons at different distances beyond the lesion epicentre of the ON. *P<o 0001 Scale bars = l00j.tm. Anterograde FIGs. 3F-3H show Rhodamine B labelling after treatment with Peni-C6DN+siCASP to confirm regenerating RGC axons in the distal ON stump (examples shown by arrowheads) immediately past the 15 lesion site (3 F), at 1000Mm (FG) and at 2000Mm (3H) from the lesion site.
FIGs. 4A-4P show the effects of intravitreal delivery of optimised Pen l-C6DN alone or in combination with siCASP2 on activation of retinal astrocytes and Muller cells and increase of CNTF in treated eyes. At 21 days after ONC and treatment with C6DN alone or in combination with siCASP2, eyes were processed for immunohistochemistry. FIGs. 4A-4L 20 show representative images of the retinae of Pen I, Pen!-C6DN and Pen I+siCASP2 treated eyes, demonstrating GFAP activated astrocytes and Muller cells with associated CNTF.
Scale bar = 100pm. FIG. 4M is a bar graph showing the effect of Penl-CÔDN alone or in combination with siCASP2 on number of activated Muller cells in the retina, Combined Pen l-CÔDN+siCASP2 treated eyes showed the highest numbers of activated 25 astrocytes/Iviuller cells and CNTF localisation in the retina. FIGs. 4N and 40 show immunolocalisation of CNTF in f3ITI-tubulin RGC in Peni (4N) and Peni-C6DN+siCASP2-treated retinae (40). Scale bar = SOpm. FIG. 4P is a bar graph showing levels of CNTF in eyes treated with combined Penl-CoDN+siCASP2. ELISA detected increased levels of CNTF in eyes treated with combined Penl-C6DN+siCASP2. ***P<0 0001 30 FIGs. SA-SI show the effects of intravitreal delivery of optimised Penl-C6DN alone or in combination with siCASP2 on adult retinal cultures prepared 5 days after ONC. Intact retinal cultures did not contain GFAP glia (FIGs. 5A, SI), while increasing numbers of GFAW glia were observed in retina dissociated after 5 days and untreated (FIGs. SB, SI) or with intravitrea treatment of Peni (FIGs. SC, SI), Penl-C6DN (FIGs. SD, SI), Penl-siCASP2 (FIGs. SE, SI) arid Penl-C6DN+siCASP2 (FIGs. SF, SI). Retinal cell cultures prepared from Penl-CÔDN+siCASP2-treated eyes that have also had suppression of CNTF signalling with either the MAB228 (Sg/eye) (FIGs. SG, SI) or the JAKISTAT pathway inhibitor AG490 5 (17mM/eye) (FIGs. SH, 51), contain significantly reduced numbers of GFAP glia to the baseline levels observed after ONC or ONC+Penl treatment. The numbers of GFAP glia positively correlated with the levels of CNTF production in culture, while treatment with MAB228 or AG490 suppressed CNTF production to baseline levels. ***<QQQffl, Scale bar = SOpm. 10 FIGs. 6A-6L show the effects of intravitreal delivery of optimised Pen 1-C6DN alone or in combination with siCASP2 on adult retinal cultures prepared 5 days after ONC on neurite outgrowth. FIGs. 6A-611 show neurite growth in culture. In untreated cultures (FIG. 6A) and cultures prepared 5 days after Peni vehicle treatment (FIG. 6B), few if any RGC grew neurites, while retinal cultures prepared from animals treated with Penl-CNTF (1.5g/eye) 15 (FIG. 6C) increased the mean neurite length (FIG. 61), mean number of RGC with neurites (FIG. 6J) and RGC survival (FIG. 6K). In retinal cultures prepared from eyes treated with PenI-C6DN and Penl-siCASP2 (FIGs. 6E, 61-K), similarly increased mean neurite length and number of RGC with neurites were observed, while RGC survival was 55% and 98%, respectively. In retinal cultures prepared from animals treated with Pen l-C6DN+siCASP2 20 (FIGs. 6F, 61-IC), significantly more neurite outgrowth was observed in terms of RGC neurite length and the number of RGC with neurites, while RGC neuroprotection was also 98%, However, simultaneous treatment with either MAB228 (FIGS. 6G, 61-K) or AG490 (FIGs.
6H, 61-K) in Penl-CÔDN+siCASP2-treated cultures abrogated RGC neurite outgrowth without affecting RGC viability. ELISA from cells treatment with Peni-C6DN+siCASP2 25 (FIG. 6L) showed significantly high levels of CNTF compared to those treated with CNTF or other monotherapies. **P<o 001 ***P<o 0001 Scale bar = 4Opm.
FIGs. 7A-7H show ii; viva that Pen 1-C6DN+siCASP2-stimulated RGC axon regeneration is abrogated by MAB228 and AG490, without affecting RGC survival. After ONC, animals were intravitreally injected with optimised Peni-C6DN+siCASP2 and rat IgG (Sug/eye), 30 MAB22S (Sug/eye) and AG490 (17mM/eye). RGC survival was quantified by FG counting in retinal wholemounts and showed that blocking Penl-C6DN+siCASP2-mediated RGC axon growth by MAB228 or AG490 did not impact on RGC survival (FIGs, 7A-7D) but did block RGC axon regeneration (FIGs. 7E-7H), *P<o 0001 compared to PenI-CoDN+siCASP2+MAB228 or Penl-C6DN+siCASP2+AG490, Scale bars in FIGs. 7A-7C = SORm; scale bars in FIGs. 7E-7G and insets (i) and (ii) = IOOtm.
FIGs. 8A-8l show that blocking Pen]-CÔDN+siCASP2-stimulated RGC axon regeneration by MAB228 and AG490 also blocks (A-J) glial activation (FIGs. 8A-SJ) and reduces CNTF 5 production in the eye (FIG. 8K). ***P<o 0001 Scale bar = SOpm. GCL = ganglion cell layer, INL = inner nuclear layer, ONL = outer nuclear layer.
FIGs. 9A-9C provide a schematic representation of a proposed mechanism of CNTF-induced gliosis in the retina after ONC and Penl-C6DN+siCASP2 treatment. After ONC, the low levels of injury-induced CNTF bind to the CNTF receptor and sub-optimally activate the 10 JAK/STAT pathway and hence little or no axon regeneration occurs (FIG. 9A). After ONC and intravitreal delivery of Penl-CÔDN+siCASP2 however, glial activation occurs in response to both injury and the presence of Peni-C6DN+siCASP2 (FIG. 9B). This gliosis leads to CNTF release, which further stimulates reactive gliosis in an autocrine manner, enhancing CNTF release. In addition, treatment with Penl-C6DN+siCASP2 also promotes 15 release of proinflammatory cytokines from microglia that stimulate further release of CNTF from astrocytes and Muller cells. These high litres of CNTF activate the CNTF receptor and promote significant RGC axon regeneration through the JAK/STAT pathway. In contrast, blocking the gpl3O component of the CNTF receptor with MAB228 or blocking the JAKISTAT signalling pathway with AG490 blocks the autocrine response, suppresses 20 proinfiammatory cytokine-induced CNTF release leading to reduced CNTF titres, depressed reactive gliosis and hence RGC axon regeneration is prevented (FIG. 9C).
FIGs bA-bE shows the effect of suppression of caspase-2 and caspase-6 on microglial activation in the retina, Immunohistochemistry demonstrates low levels of immunoreactive OX-42 in retinal microglia after ONC in (A) Pent controls, (B) in Penl+siCASP2 and (C) 25 PenI-C6DN-treated retinae. Higher levels of OX-42 immunoreactivity are observed after combined Penl-CÔDN+siCASP2 treatment (D), with the microglia showing an activated morphology. Quantification of the pixel intensities reflect these changes and demonstrate the significantly elevated levels of OX-42 immunoreactivity in Penl-C6DN+siCASP2-treated retinae(E). ***p<0 000] Scale bar= 5ORm. 30
DESCRIPTION OF SOME EMBODIMENTS OF THE INVENTION
The invention, in some embodiments thereof, relates to use of agents for inhibition of caspase-2 and caspase-6 in protection of retinal neuronal cells from apoptosis and promotion of axon regeneration.
The present inventors have shown that combined inhibition of caspase-2 and inhibition of 5 caspase-6 activates astrocytes and Muller cells, increases ciliary neurotrophic factor (CNTF) levels in the retina and leads to enhanced ROC axon regeneration.
The present inventors delivered an inhibitor of caspase-6 (caspase-6 dominant negative (C6DN), coupled to a cell penetrating peptide, Penetratin-] (Peni) either alone or in combination with an inhibitor of caspase-2 (chemically stabilised siRNA to caspase-2, 10 5iCASP2) and measured the effects on RGC neuroprotection and axon regeneration. It was shown that caspase-2 inhibition provided significantly more RGC neuroprotection after ONC than caspase-6 inhibition. Moreover, the combination of C6DN and siCASP2 did not potentiate ROC survival over that observed with siCASP2 alone. However, when both caspases were simultaneously inhibited, RGC axon regeneration was indirectly and 15 significantly increased through activation of retinal glial-dependent CNTF secretion.
In dissociated adult rat mixed retinal cultures, C6DN+siCASP2 treatment also significantly increased GFAP glial activation, increased the expression of CNTF in culture, and subsequently increased the number of ROC with neurites and the mean ROC neurite length.
These effects were abrogated by the addition of MAB228 (a monoclonal antibody targeted to 20 the gpi3O component of the CNTF receptor) and AG490 (an inhibitor of the JAK/STAT pathway downstream of CNTF signalling). Similarly, in the ON crush injury model, MAB228 and A0490 neutralised C6DN+siCASP2-mediated RGC axon regeneration, Muller cell activation and CNTF production in the retina without affecting RGC survival.
It is therefore concluded that axon regeneration promoted by suppression of caspase-2 and 25 caspase-6 is CNTF-dependent and mediated through the JAK/STAT signalling pathway.
These findings offer insights for the development of effective therapeutics for promoting RGC survival and axon regeneration.
The principles, uses and implementations of the teachings herein may be better understood with reference to the accompanying description and figures. Upon perusal of the description 30 and figures present herein, one skilled in the art is able to implement the invention without undue effort or experimentation.
Before explaining at least one embodiment in detail, it is to be understood that the invention is not necessarily limited in its application to the details of constmction and the arrangement of the components and/or methods set forth herein. The invention is capable of other embodiments or of being practiced or carried out in various ways. The phraseology and terminology employed herein are for descriptive purpose and should not be regarded as 5 limiting.
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable 10 subcombination or as suitable in any other described embodiment of the invention.
Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art, Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the scope of the appended claims. 15 Citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the invention.
Experimental Example t: Combined suppression of caspase-2 and caspase 6 protects retinal an1ion 20 cells from apopotosis and promoted axon rezeneration Materials and methods Optic nerve crush (ONC) All animal procedures were licensed and approved by the UK Home Office and the University of Birmingham Ethical Review Committee, Adult, female 6-8 week-old Sprague-25 Dawley rats (1 80-220g) were anaesthetised with Isofluorane inhalation anaesthesia (Janssen Pharmaceuticals, Oxford, UK), the ON was exposed through a supra-orbital approach and cmshed bilaterally 2 mm from the globe using watchmaker's forceps as previously described by the present inventors (Vigneswara et, al,, 20t2), None of the animals developed cataracts, confirming that the lens had not been injured during surgery, 30 Penl-C6DN The mutant Caspase-6 (Cysi63Ala) dominant negative (C6DN) expression construct was a kind gift of G. S. Salvesen, Sanford-Bumham Institute, La Jolla, CA, C6DN was purified in as described by Denault and Salvensen, 2003. Penetratin-(Pen-I) was custom synthesized by PolyPeptide Laboratories, Torrance, CA. Pen-I and C6DN were linked by incubating 5 equimolar amounts at 37°C for 24 hours to generate disulfide bonds. Linkage was confirmed by non-reducing 20% PAGE with western blotting using anti-His antibodies, s1CASP2 dsNA to CASP2 (5iCASP2, also identified as QPI-1007) was provided by Quark 10 Pharmaceuticals Inc., Nes Ziona, Israel.
siCASP2 (QPI-1007) is a double-stranded nucleic acid compound having the structure: 5' iB -GCCAGAAUGIJGGAACUCCU 3' (sense strand) 3' CGGUCUUACACCUUGAGGA 5' (antisense strand) wherein each A, C, U, and G is a ribonucleotide and each consecutive ribonucleotide is 15 joined to the next ribonucleotide by a phosphodiester bond; wherein the sense strand comprises, counting from the 5' tenninus, an unmodified ribonucleotide at positions 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 and 19, a L-deoxycytidine at position 18, and an inverted deoxyabasic moiety (iB) 5' cap; and wherein the antisense strand comprises, counting from the 5' terminus, a 2'-O-Methyl sugar modified ribonucleotide at positions 2, 20 4, 6, 8, II, 13, 15, 17 and 19 and an unmodified ribonucleotide at positions 1, 3, 5, 7, 9, 0, 12, 14, 16 and 18.
in viva experiments In the preliminary Penl-C6DN dose-finding experiment, groups comprised six rats/treatment 25 (i.e. 12 eyes/treatment): (I) Intact, (2) ONC+Penl (vehicle control) (PenI, OpM C6DN), (3) 2j.dvl Penl-C6DN, (4), 4pM Penl-C6DN, (5), 511M Penl-C6DN and (6) 7pltvI Penl-C6DN.
To monitor caspase-2 and caspase-6 activation over the first 7 days, groups of six rats/treatment (12 eyes/treatment) were killed at 4 and 7 days after ONC, while a further three rats (6 eyes) were used as intact controls, In further experiments, groups of six rats/treatment 30 (12 eyes/treatment) were used to determine the effects of siCASP2 and pre-optimised Pen]-C6DN on the levels of caspase-2 and cleaved Lamin A/C, a substrate for active caspase-6 and comprised: (t) ONC+Pent, (2) ONC+Penl-C6DN, (3) ONC+Penl+siCASP2 and (4) ONC+Penl -C6DN+siCASP2. To determine the effects of siCASP2 and pre-optimised Peni -C6DN singly and in combination on RGC survival, groups of six rats/treatment (t2 5 eyes/treatment) were used and groups comprised: (1) ONC+PBS, (2) ONC+Penl, (3) ONC+Penl-C6DN, (4) ONC+Penl+siCASP2 and (4) ONC+Penl-C6DN+siCASP2 and (5) Intact controls. To determine the effects of siCASP2 and pre-optimised Peni-C6DN on ROC axon regeneration and Muller cell activation in the retina six rats/treatment (12 eyes/treatment) were used and groups comprised: (1) ONC+Penl, (2) ONC+Penl-C6DN, (3) 10 ONC+Peni+siCASP2 and (4) ONC+Penl-C6DN+siCASP2, To determine the levels of CNTF in the eye, groups of six rats/treatment were used and groups comprised: (1) ONC+Penl, (2) ONC+Penl-CÔDN, (3) ONC+Pen]+siCASP2 and (4) ONC+Penl-C6DN+siCASP2. To determine the effects of MAB228 and A0490 on RGC survival, axon regeneration, retinal Muller cell activation and CN]TF localisation, groups of twelve 15 rats/treatment (6 rats for FG labelling and 6 rats for GAP -43 and GFAP/CNTF immunohistochemistry) were used and groups comprised: (1) ONC+Peni- C6DN+siCASP2+IgG, (2) ONC+Penl -C6DN+siCASP2+MAB228 and (3) ONC+Penl -C6DN+siCASP2+ AG490. Finally, to determine the levels of CNTF in the eye after treatment with MAB228 and AG490, groups of six rats/treatment were used and groups 20 comprised: (I) ONC+Penl-C6DN+siCASP2+lgG, (2) ONC+Penl-C6DN+siCASP2+MAB228 and (3) ONC+Penl -C6DN+siCASP2+AG490.
Intravitreal injections In a preliminary experiment, Penl-C6DN was titrated to determine the optimal dose required 25 to promote maximal RGC survival, Immediately after ONC, animals (n = t2 eyes/group) received Peni vehicle or increasing concentrations of CODN from 2, 4, 5 and 7pM Penl-C6DN using glass micropipettes. In further experiments 5pM of Peni or Penl-C6DN was injected. Intravitreal injections were repeated every 7 days based on previous experiments by the present inventors with other caspase inhibitors (Ahmed el at, 201 1). 2Opg/eye of 30 siCASP2 was injected along with either SpIVI Peni or Penl-C6DN. The optimal dose of MAB22S was pre-detennined by its ability to reduce CNTF levels in treated eyes, with optimal concentrations determined as 5g/eye (not shown), the inhibitor of Janus-kinase 2 (JAK) AG490 was injected at 17mM/eye (Muller ci at, 2007) and CNTF was injected at a dose of 1.5j.tg/eye (Muller ci at, 2007). All intravitreal injections were made up in a final volume of Sp.l/eye and repeated every 7 days.
Retinal wholemounts 5 At 19 days after ONC, 2pJ of 4% FluoroGold (FG; Cambridge Bioscience, Cambridge, UK) in PBS was injected into the ON, between the lamina cribrosa and the site of ONC in all experimental groups (Vigneswara ci al., 2012), Animals were killed 48h later and intracardially perfUsed with 4% formaldehyde (TAAB Laboratories, Aldermaston, UK) in PBS. Retinae were harvested and flat mounted onto Superfrost Plus microscope slides (VWR 10 International, Lutterworth, UK). Retinal wholemounts were dried onto glass slides and mounted in Vectamount (Vector Laboratories, Peterborough, UK). Samples were then randomised by a second investigator and photographs were captured using a Zeiss Axioplan 2 fluorescent microscope equipped with a digital Axiocam HIRc camera, controlled through Axiovision 4 software (all from Zeiss, Hertfordshire, UK). The number of FG-labelled ROC 15 was counted using the automated particle counting software in ImagePro (Version 6.0) (Media Cybernetics, Bethesda, USA) from photographs of 12 rectangular (0.36 X 0.24mm) areas/retina, 3 from each retinal quadrant, placed radially at inner (1/6 eccentricity), mid-periphery (1/2 eccentricity) and outer retina (5/6 eccentricity) from the centre of the optic disc. The number of FG-labelled RGC in the 12 images/retina were divided by the counting 20 area and resultant numbers were pooled to calculate mean densities of EG-labelled RGC/mm2 for each retina (Peinado-Ramon ci at, 1996, Vigneswara ci at, 2012).
Anterograde labelling of regenerating RGC axons with Rhodamine B isothiocyanate Anterograde labelling of regenerating axons using Rhodamine B isothiocyanate (RITC; 25 Sigma, Poole, UK) was performed as described previously (Thanos ciat, 1987). Briefly, Sjil of a 2.5% solution of RITC was intravitreally injected using glass micropippettes 2 days prior to sacrifice. Animals were killed by overdose of CO2, perfused with 4% formaldehyde in PBS and the ON were prepared for cryo-sectioning as described below, Tissue preparation and sectioning After intracardiac perthsion with 4% formaldehyde in PBS, eyes and ON were removed and prepared as described previously (Douglas el at, 2009, Ahmed c/ at, 2010, Vigneswara ci at, 2012). Briefly, eyes and ON were post-fixed in 4% formaldehyde (TAAB) in PBS, incubated in a graded series of sucrose solutions in PBS and then embedded in OCT 5 mounting medium (Raymond A Lamb Ltd) prior to freezing at -80°C. Later, 15r.tm thick parasaggital and longitudinal sections of eye and ON, respectively, were cut on a cryostat (Bright Instruments, l-luntingdon, UK), adhered onto glass slides and stored at -20°C until required.
Immunohistochemistry Immunohistochemistry was performed on sections of retina and ON as described previously (Douglas et at, 2009, Ahmed ci at, 2010, Ahmed ci at, 2011, Vigneswara ci at, 2012).
Briefly, sections were washed in PBS and non-specific binding was blocked for 20 mm before incubation with the relevant primary antibody. Monoclonal anti-GAP43 (1:500 15 dilution in PBS containing 3% BSA and 0,05% Tween-20; Invitrogen, Paisley, UK) was used to localise regenerating axons; monoclonal anti-GFAP and a polyclonal anti rat-CNTF (1:500; Promega, Southampton, UK) was used to stain for astrocytes and CNTF in retinal sections and in retinal cultures; monoclonal anti-f3111-tubulin (1:200; Sigma, Poole, UK) was used to stain for ROC and their neurites in retinal cultures. Sections were washed in PBS and 20 incubated with appropriate Alexa Fluor 488 and Texas Red-labelled secondary antibody (Invitrogen) for t hour at room temperature, washed, mounted in Vectashield mounting medium with DAPI (Vector Laboratories) and examined under an Axioplan-2 epi-fluorescent microscope (Zeiss).
Quantification of Muller cell processes Retinal Muller cell activation was quantified as described previously (Ahmed ci at, 2010), Briefly, after GFAP immunohistochemistry, GFAP Muller cell processes were counted along a 250r.tm horizontal line in retinal sections, placed orthogonal to the radial plane through the middle of the internal plexiform layer. The mean GFAP cell counts for each 30 condition (n = 12 retinal sections/condition) were calculated and expressed as mean +SEM, Adult rat retinal cultures Five days after ONC and intravitreal treatments of pre-optimised reagents (concentrations detailed above), animals (n = 6 eyes/group) were killed by CO2 overdose, retinae harvested and dissociated using a Papain dissociation system following the manufacturer's instructions (Worthington Biochemical, Lakewood, NJ, LISA), as described previously (Vigneswara et 5 at, 2013). Retinal cells from untreated rats were also prepared in a similar way. Retinal cells were plated at a density of 125 x 1 03/well into 8-well chamber slides pie-coated with laminin and poly-D-lysine and cultured in 300j.tl Neurobasal-A medium supplemented with B27 supplement and gentimicin (all from Invitrogen, Paisley, UK). Cells were cultured for 4 days at 37°C and 5% CO2 before either removal of culture medium and cells for ELISA (see below 10 for description) or fixation in 4?'b formaldehyde in PBS for immunocytochemistry.
Experiments comprised of n = 3 wells/treatment and each experiment was repeated on 3 separate occasions and hence results for each data point are the mean +SEM from 9 wells/treatment.
Immunocytochemistry Fixed cells were immunostained for GFAP and f3111-tubulin to determine the number of GFAP astrocytes and Muller cells and to measure the number of RGC with neurites and the neurite lengths, as described previously (Ahmed et a)., 2010). Briefly, cells were washed in PBS, permeabilised and blocked using PBS containing 3% BSA and 0.1% Triton X-100 for 20 mm at room temperature before incubation with the mouse anti-GFAP (1:500 dilution, Sigma, Poole, UK) or mouse anti-13111-tubulin antibodies (1:200 dilution, Sigma) for 1 hour at room temperature in a humidified chamber. Cells were then washed in PBS and incubated for 1 hour at room temperature with Alexa-488 anti-mouse IgG (1:400 dilution; Invitrogen).
After further washes in PBS, sections were mounted under coverslips using Vectamount 25 containing DAPI (Vector Laboratories, Peterborough, UK) and viewed under a Zeiss Axioplan 2 fluorescent microscope equipped with an Axiocam HRc and Axiovision software, as described earlier, Immunocytochemistry controls with primary antibody omitted were included in each mn and the negative control slides were used to set the baclcground threshold levels for non-specific staining (not shown) during image capture. 30 RGC survival and neurite outgrowth and quantification of astrocyte activation The mean number of surviving III-mbu1in RGC, mean neurite length, the number of RGC with neurites and the number of GFAP astrocytes/Muller cells were quantified as described previously (Vigneswara et at, 2013), Briefly, each anonymised chamber slide was divided into 9 quadrants and images of ROC, their neurites and GFAP astrocvtesA'tuller cells were 5 captured randomly from each quadrant. Axiovision (Zeiss) was then used to measure the neurite lengths, while TmagePro (Version 6.3; Media Cybernetics, Bethesda, MD, USA) was used to quantify the number of 13111-tubulin RGC with neurites longer than the RGC diameter and the number of GFAP astrocytes/Muller cells. Neurite outgrowth from at least RGC/treatment was measured, except in untreated and Peni -vehicle treated cultures in 10 which 100 RGC were assessed (i.e. all RGC that grew neurites longer than the RGC diameter).
ELISA for CNTF For detection of CNTF in vitro, cultured cells and culture medium were homogenised in cell 15 lysis buffer and clarified by centrifugation. Lysates were then assayed for CNTF and compared to culture medium only conditions to account for any background CNTF. Retinae harvested from in vivo experiments were homogenised in cell lysis buffer, clarified by centrifugation and the supematant was frozen at -20°C until required for assay. A commercially available rat CNTF ELISA kit (R&D Systems, Oxford, UK) was used to detect 20 CNTF in cultured retinal cell lysates, following the manufacturer's instructions.
Protein extraction and Western blotting Six rats (12 eyes/treatment) were killed by an overdose of CO2 and total protein from retinae and where appropriate, ON, were extracted in cell lysis buffer and processed for Western 25 blotting as previously described (Ahmed ci at, 2005, Ahmed et at, 2006, Ahmed ci at, 2010). Western blots were probed overnight at 4°C with antibodies against: rabbit anti-human caspase-2 (CASP2; Abcam); rabbit anti-Lamin A/C and rabbit anti-human GAPDH, all from Cell Signalling Technology (Danvers, MA, USA). Relevant protein bands were detected with an appropriate FIRP-labelled secondary antibody (GE Healthcare, Buckinghamshire, UK) and 30 detected using an enhanced chemiluminescence system (ECL) (GE Healthcare). Blots were stripped and re-probed as required.
Densitometry Western blots were quantified by densitometry as described previously (Ahmed ci at, 2006, Douglas dat, 2009, Ahmed el at, 2010). Briefly, blots were scanned into Adobe Photoshop and TIFF files were analysed in Scionimage (version 4.0.2, Scion Corp. Maryland, USA) using the built-in gel plotting macros. The integrated density of each band of interest in each 5 lane was calculated for 3 separate blots from 3 independent experiments.
Statistical analysis The significance of differences between sample means were calculated using GraphPad Prism (GraphPad Sofiwa.re Inc., Version 4.0, CA, San Diego, USA) by one-way analysis of 10 variance (ANOVA) followed by pact-hoc testing with Dunnett's method.
Res tilts Characterization of a specific inhibitor of caspase-6 To determine the functional relevance of the up-regulation of cleaved caspase-6 (C-CASP6) 15 expression afler ONC, caspase-6 activity was inhibited in the retina. A previously characterized C6DN construct (Edgington et at, 2012) was used to block activation/activity of caspase-6. For access into the retina, C6DN was disulphide-linked to Penetratin-1 (Peni), a cell penetrating peptide (Davidson ci at, 2004), the disulphide bond was broken by the reducing environment of the cell cytoplasm, thus releasing the peptide cargo and allowing it 20 to act at its cellular target.
The neuroprotective efficacy of this construct was examined by assessing RGC survival after ONC and intravitreal delivery of a range of doses ofPenl-C6DN, by counting the number of FluoroGold (FO) backlabelled ROC in retinal wholemounts. Compared to the vehicle treatment (Pen I), which left 400 ± 45 FG-labelled RGC/mm2, Peni-C6DN (2, 4 and SRM) 25 caused a dose-dependent statistically significant increase in the numbers of surviving FO-labelled RUC, to a maximal level of 987 + 65 RGC/mm2 (Fig. IA-F). Concentrations of Penl-C6DN above 5pM did not significantly increase the number of FG-labelled ROC; thus, maximal RGC protection (60%) was observed with 5pM PenI-C6DN, These resuhs demonstrated that specific blockade of caspase-6 activity, using Pen l-C6DN, significantly 30 enhances RGC survival.
To ensure that each intervention affected only the targeted caspase, the expression of caspase- 2 was examined in retinae treated with Penl and Penl-C6DN by western blot. No decrease in caspase-2 levels was detected (Figs. 2A and 2B). Peni-C6DN decreased basal cleaved Lamin AIC (a substrate of caspase-6 (Orth ci at, 1996, Takahashi ci at., 1996, Ruchaud ct cii., 2002, Mintzer ci at, 2012)) levels while, in Pen! and Pen I+siCASP2-treated retinae, basal cleaved 5 Lamin A/C levels were unaffected (Fig. 2A and 2B), indicating constitutive caspase-6 activity in non-RGC cells. These results demonstrated that the caspase-2 and caspase-6-specific inhibitors disclosed herein do not have cross reactivity but that each specifically regulates their targeted caspase.
To assess the neuroprotective properties of the combined Pent-C6DN and siCASP2, optimal 10 doses of Pen I -C6DN were intravitreally inj ected together with previously optimi sed dose of siCASP2 (Ahmed ci at, 20]]) after ONC, The number of FluoroGold backfilled surviving ROC were assessed at 21 days after ONC. In eyes treated with the vehicle control (Penl), 405 + 34 RGC/mm2 remained at 21 days after ONC (Figs. 2C and 2D), while in Pen t-C6DN treated eyes 979 + 34 RGC/mm2 remained (Figs. 2C and 2D). However, Penl+siCASP2 15 protected 2145 ± 54 RGC/mm2, while Penl-C6DN+siCASP2 did not significantly improve RGC neuroprotection over that observed for siCASP2 alone (Figs. 2C and 2D), Compared to intact controls, Penl-C6DN promoted 60% RGC neuroprotection while siCASP2 alone or Penl-C6DN+siCASP2 protected >95% of RGC from apoptosis at 21 days after ONC, These results suggest that both Pen]+siCASP2 and Pen]-CGDN+siCASP2 promote optimal RGC 20 survival.
Penl-C6DN+siCASP2 promoted RGC axon regeneration Previous studies have suggested that active caspase-2 and caspase-6 modulate different signalling pathways. The present inventors therefore tested whether simultaneous blocking of 25 these caspases using a combined treatment of Pen] -CGDN and siCASP2 would promote RGC axonal regeneration. It has been previously shown that siCASP2 decreased caspase-2 expression by almost 90% after ONC but there was little associated RGC axonal regeneration (Ahmed dat, 201]), In other models of axonal degeneration (trophic factor deprivation and stroke) caspase-6 activation initiates axonal degeneration (Nikolaev ci at, 2009, Akpan ci at, 30 20!!). To determine whether caspase-6 might have a similar function in RGC degeneration, caspase-6 function was inhibited using C6DN and combined with delivery of siCASP2 to promote optimal ROC survival and thus enhance the possibility of reduced RGC loss and axon degeneration. At 7 and 14 days afier ONC, animals received intravitreal injections of siCASP2 and/or Pen 1-C6DN, and analysed RGC axon regeneration was analyzed through the crush site at 21 d after ONC using antibodies against GAP-43, a marker for regenerating axons (Berry e. at, 1996, Leone/a!., 2000). 5 When the ON of Peni treated animals were sectioned longitudinally and immunostained for GAP-43 (Figs. 3A and 3E), few GAP43 axons were seen in the proximal ON segment, and none traversed the lesion site to enter the distal ON segment. In Penl-C6DN treated ON (Figs. 3B and 3E), more GAP43 axons occupied the proximal ON but few traversed the lesion site, although some of these penetrated into the distal ON up to 1500i,tm from the 10 lesion centre, The mean number of axons/ON were low after the monotherapies, so that the numbers of GAP4f axons were similar in the Penl+siCASP2-treated and Penl-C6DN-treated groups, with only occasional axons penetrating >1000l1m from the lesion centre and into the distal ON (Figs. 3C and 3E). The greatest numbers of GAP43 axons were present in the Pen L.CÔDN+siCASP2-treated groups, with 95 ± 9 and 72 ± 8 axons/section growing at 15 1000im and 2500p,m, respectively, from the lesion (Figs. 3D and 3E). These results suggest that, while Pen l-C6DN or siCASP2 alone promote little ROC axon regeneration, combined delivery ofPenl-C6DN+siCASP2 promotes significant ROC axon regeneration, Anterograde labelling with Rhodamine B isothiocyanate confirmed similar numbers of regenerating RGC axons emerging from the lesion site and growing through the distal optic 20 nerve stump (arrowheads) to 1000 and 2üü0m from the lesion site (Fig 3F-H), For example, the number of RITC-labelled axons at 250, 1000 and 2000pm from the lesion site were, 200 + 14, 210 + 12 and 30 + 10 axons/section, respectively (not illustrated).
Penl-C6DN+siCASP2 delivery enhanced retinal glia activation and upregulated CNTF 25 production in glia and occasional RGC Studies of retinal glial activation in models of RGC axon regeneration have shown that glial activation is correlated with axon regeneration (Berry c/at, 1996, Leon c/cL, 2000, Lorber et at, 2002, Yin et at, 2003, Lorber eta!., 2005, Pernet and Di Polo, 2006, Muller et at, 2007, Lorber c/al., 2008, Lorber c/at, 200c, Ahmed c/at, 2010, Lorber c/at, 2012), It has 30 also been shown that after ONC, inflammation-mediated glial activation in either the vitreous or in the ON injury site promoted significant RGC survival, but that only vitreal inflammation with associated retinal glial activation was correlated with axon regeneration, This observation suggests that activated retinal glia secrete factors that are conducive for RUC axon regeneration (Ahmed eta!., 2010). Moreover, activated retinal astrocvtes express CNTF in response to lens injury or intravitreal Zymosan injections; the downstream JAKISTAT3 pathway is strongly activated in regenerating RGC and the lens injury-induced 5 switch of RGC to a regenerative state is dependent on CNTF and JAK/STAT3 signalling (Muller et a&, 2007). Thus, it was investigated whether intravitreal delivery of Peni-C6DN+siCASP2 after ONC also promoted retinal glial activation and CNTF expression.
GFAP astrocytes/Muller cell end-feet were similarly activated and low levels of CNTF appeared in the nerve fibre layer (NFL) after ONC in vehicle control Pent-treated (Figs. 4A-10 4C) and in Pen]-C6DN-treated eyes (Figs. 4D-4E). In Pen+siCASP2-treated eyes (Figs. 4G- 41), more GFAP astrocytes/MUller cells and CNTF were observed in the NFL compared to Peni and Pen 1-CoDN treated eyes. The greatest levels of GFAP and CNTF staining were observed in Pent-C6DN+siCASP2-treated eyes, with more numerous GFAP Muller cell processes spanning the entire radial width of the retina (Figs. 4J-4L), reflected by twice the 15 number of activated Muller cell processes counted in these eyes compared to the numbers in other treatments (Fig. 4M). Tn addition, occasional RGC were immunopositive for CNTF only after combined suppression of caspase-2 and -6, suggesting that some RGC may also respond to the effects of the combined treatments (Figs. 4N and 40).
ELISA confirmed higher titres of CNTF in the retina of eyes treated with PenT-20 C6DN+siCASP2 than that measured in the eyes from any other treatments (Fig. 4P).
Together, these results suggest that the bi-therapy of Pen]-C6DN+siCASP2 activates retinal astrocytes/Muller cells to produce high titres of CNTF and that this may explain the enhanced RGC axon regeneration observed after this combined treatment.
Another possible mechanism of the enhanced CNTF production by retinal glia and ROC was 25 investigated, since caspase-6 suppression can up-regulate proinflammatory cytokine release by microglia that may then facilitate enhanced CJNI'F release from activated retinal cells.
Immunohistochemistry for OX-42, a marker of microglia, demonstrated higher levels of microglial activation after caspase-2 and caspase-6 monotherapy suppression, and that combined suppression of caspase-2 and caspase-6 synergistically increased this microglial 30 activation (Figs. bA-TOE). These results suggest that microglial activation and subsequent proinflammatory cytokine production may contribute to the activation of retinal glia and subsequent CNTF release.
RGC cultures prepared from ONC+Penl-C6DN'+siCASP2 show enhanced glial activation and significant titres of CNTF in culture medium Many of the responses observed in viva after ONC and treatment may be studied in vitro using adult retinal cultures. To further assess whether Pen]-C6DN+siCASP2 promoted glial activation and indirectly enhanced neurite outgrowth of surviving RGC, retinal cultures from 5 intact untreated eyes and from eyes 5 days after ONC and treatment with Peni vehicle, Penl-C6DN, Pen I+siCASP2 and Pen l-C6DN+siCASP2 treatment were prepared. After 3 days in culture, there were no activated GFAP glia detected in retinal cell cultures derived from untreated eyes (Figs. 5A and 51). In retinal cell cultures prepared after ONC (Fig. SB and I) and ONC followed by Pent treatment (Fig. SC and I), 1396 + 259 and 1367 + 379 activated 10 GFAP glia were present, respectively. Cultures prepared from eyes after ONC and mono-therapy with Pen]-C6DN and Penl+siCASP2, contained enhanced numbers of activated GFAP glia (2867 + 379 and 2433 + 208, Figs. SD, SE and SI), respectively, whilst cultures prepared from Pent-C6DN+siCASP2-treated eyes contained the greatest numbers of GFAP glia (4900 + 355 glia, Figs. SF and SI). These results demonstrated that PenI-15 C6DN+siCASP2 treatment led to the survival of enhanced numbers of activated GFAP glia in retinal cell cultures and this confirmed the in viva findings.
Since reactive astrocyte gliosis can be induced by elevated levels of CNTF (Winter et aL, 1995, Escartin et aL, 2007, Kirsch et at, 2010) the present inventors investigated whether blockade of the gpl3O signalling component of the CNFF receptor complex using MAB228 20 or of the JAK/STAT3 pathway using AG490 suppressed the Penl-C6DN+siCASP2-mediated survival of activated glia in retinal cell cultures, In cultures prepared from eyes after ONC and treatment for 5 days with MAB228 and AG490, it was observed that the Peni-C6DN+siCASP2-mediated enhancement of GFAP glia numbers in retinal cultures was depressed to control ONC and ONC+Penl levels (Figs. SC, SH and SI, respectively) by the 25 inhibitors of CNTF signalling.
Since high levels of CNI'F were observed after ONC and treatment with Peni-CÔDN+siCASP2 in viva, ELISA was used to monitor CNTF titres in defined cell culture medium after treatment, It was demonstrated that in the media of cultures prepared from untreated, ONC-and Pen I-treated retinae, low levels of CNTF were detected (Fig, Si) but, in 30 cultures from both Penl-C6DN and Penl+siCASP2-treated retinae, approximately 38 + 8 ng/mg of CNTF protein were present in the media. The levels of released CNTF were 3-fold higher in cultures prepared from retinae after Penl-C6DN+siCASP2 bi-therapy (115 + 12 ng/mg of protein). However, addition of the inhibitors of CNTF signalling, MAB22S or A0490, depressed the numbers of Penl-C6DN+siCASP2 activated glia and the released CNTF levels to those observed in the cultures from control ONC and Pent-treated eyes.
These results suggested that Penl-C6DN+siCASP2 activates retinal glia and CNTF production, while blockade of CNTF signalling not only suppresses the numbers of activated 5 glia but also CNTF production in culture.
Treatment of retinal cell cultures with PenI-C6DN+siCASP2 promotes RGC neurite outgrowth Since Penl-C6DN+siCASP2 treatment activates retinal glia and secretion of CNTF in vivo 10 and in vitro, it was hypothesized that RGC survival and neurite outgrowth would also be enhanced in these cultures. The present inventors therefore investigated whether Peni-C6DN+siCASP2 bi-therapy led to stimulated RGC survival and neurite outgrowth in retinal cell cultures, and whether MAB228 and AG490 treatment impacted on the ROC survival and neurite outgrowth. In retinal cultures from untreated (Figs. 6A and 61-6K) and PenI-treated 15 eyes (Figs, 6A and 61-61K), very few RGC survived and grew neurites. After Pent+CNTF treatment, RGC survival was increased by 1.7-fold, while the number of RGC with neurites increased to 25 + S RGC with mean neurite lengths of 100 + i0m recorded, compared to PenI alone (Figs. 6B and 61-6K). The addition of Pen]-C6DN (Figs. 6C, 61 and 6J) and Penl+siCASP2 on their own (Figs. 6D, 61 and 6J) promoted 50% fewer RGC with neurites 20 (1012) than that seen with CNTF, with RGC neurite lengths of between 45-SOj,tm. RGC survival was similar in Penl-C6DN and Penl+CNTF treatment groups, while Penl+siCASP2 provided greater than 2-fold more RGC survival than CNTF or Pent-C6DN (Fig. 6K).
Combined Pen]-C6DN+siCASP2 treatment significantly increased the number of RLIC with neurites and the mean neurite length to 130 + 8 tm (Figs. 6E and 61) compared to CNTF 25 (Figs. 61 and 6J). RGC survival in the Pent-C6DN+siCASP2 and Penl+siCASP2 groups was similar, but was significantly greater than that seen with the CNTF, Penl-CÔDN or control groups (Fig. 6K). Despite the observation that RGC neurite outgrowth was completely suppressed to basal levels in Peni-C6DN+siCASP2 cultures treated with MAB228 and AG490 (Figs. 6G-6J), ROC survival remained at the same high levels as other groups 30 containing siCASP2 (Fig. 6K). ELISA to detect the levels of CNTF present in cukure media from the different treatment groups showed that cultures prepared from eyes treated with Penl-CNTF contained 23 + 3ng/ml, whilst those treated with Penl-C6DN and Pen] +siCASP2 contained 20 ± 3 and 16 ± 2.5ng/ml (Fig. 6L). However, the highest levels of CNTF (64 + 5.Sng/ml) were detected in cultures prepared from Penl-C6DN+siCASP2-treated eyes, equating to 3-fold more CNTF than after treatment with Pent-CNTF. These results suggest that RGC survival is maximal in the presence of siCASP2, while C6DN+siCASP2-mediated glial activation and subsequent high titres of CNTF secretion 5 enhances RGC neurite outgrowth.
Blocking gpI3O or the JAK/STAT pathway blocked PenI-C6DN+siCASP2-mediated RGC axon growth and suppressed glial activation and CNTF production in the retina Since suppression of gpl3O and the JAK/STAT pathway blocked Penl-C6DN+siCASP2-10 mediated RGC neurite outgrowth in vitro without affecting RGC survival, it was considered whether MAB228 and A0490 could also attenuate Peni-C6DN+siCASP2-induced ROC axon growth in vivo and assessed the impact of these treatments on RGC survival after ONC.
It was shown that after intravitreal injection of Penl-C6DN+siCASP2, ROC survival was unaffected by Iv1AB228 or A0490 treatment and remained at the same high levels as 15 treatment with a control IgG (Figs. 7A-7D). ROC axon regeneration in Peni-C6DN+siCASP2+IgG control treatment group was the same as that observed earlier (comparing Figs. 7E and 7H with Fig. 3). However, the Pent-C6DN+siCASP2-stimulated RGC axon regeneration was almost completely blocked by intravitreal injection of either MAB228 (Figs. 7F and 7H) or AG490 (Figs. 70 and 7H), suggesting that the CNTF pathway 20 was primarily involved in Pen l-C6DN+siCASP2-mediated RGC axon regeneration.
The present inventors then investigated whether MAB228 and AG490 also blocked Peni-CÔDN+siCASP2-mediated glial activation and CNTF production in the retina. After intravitreal Penl-C6DN+siCASP2 plus control IgG injections, abundant GFAP and CNTF staining was observed in astrocytes of the INFL and Muller cell radial processes (Figs. 8A-25 8C). However, in PenL-C6DN+siCASP2+MAB228 (Figs. 8D-8F) and PenI-C6DN+siCASP2+AG490-treated eyes (Figs. 80-81), GFAP and CNTF staining was markedly attenuated. The mean number of activated Muller glia processes in Peni-C6DN+siCASP+lgG treated retinae was 210 ± 5mm' compared to only 45 ± 5 and 8 ± 3 processes/mm" in Pen 1 -C6DN+siCASP2+MAB228 and Pen 1 -C6DN+siCASP2+AG490, 30 respectively (Fig. 8J). ELISA to quantify the levels of CNTF in Pen 1-C6DN-siCASP2+IgG-treated retinae measured 223 + S ng/mg of CNTF but the growth factor was barely detectable in both Pen] -C6DN+siCASP2+ MAB22S and Pen I -CÔDN+siCASP2+AG490-treated retinae (Fig. 8K).
Taken together, these results suggest that Pen]-C6DN+siCASP2-mediated RGC axon regeneration is suppressed when gp130 and JAK/STAT are blocked, and this correlates with reduced retinal glial activation and CNTF levels. Thus, inhibition of caspase-6, while capable 5 of promoting optimum RGC survival when combined with inhibition of caspase-2, might be a promising way of indirectly enhancing RGC axon regeneration after optic nerve trauma.
Discussion The data presented here demonstrate that despite low levels of detectable caspase-6 activation 10 after ONC, inhibition of caspase-6 promoted 60% RGC survival while inhibition of caspase-2 promoted greater than 95% RGC survival. Combined inhibition of caspase-6 and caspase -2 protected the majority of RGC from death but also promoted extensive retinal gliosis and significant RGC neurite outgrowth!axon regeneration through the ON lesion site and along the distal ON, RGC axon regeneration promoted by combined suppression of caspase-6 and 15 caspase-2 was mediated by glial-derived CNTF since blockade of the CNTF receptor or inhibition of JAK!STAT signalling, suppressed the associated glial activation, blocked CNTF production, inhibited RGC axon regeneration and preserved RGC viability. Taken together, the results show that combined suppression of caspase-2 and caspase-6 is ROC neuroprotective and activates a novel indirect ROC axogenic effect mediated by caspase-20 dependent gliosis and subsequent release of CNTF and JAK/STAT signalling.
Combined caspase-6 and caspase-2 suppression did not induce stronger neuroprotection than that observed with caspase-2 suppression alone, since suggesting that RGC death is predominantly mediated by caspase-2 and not caspase-6.
The sub-optimal leve's of RGC neuroprotection seen after C6DN treatment, suggest that use 25 of an eflèctive caspase-6 inhibitor may increase RGC survival by achieving greater levels ol' caspase-6 down-regulation/inhibition. For example, C6DN suppressed Lamin A/C cleavage, a primary target of caspase-6 activity, by on'y 60% compared to treatment groups that did not contain C6DN (see Fig. 2A). Greater levels of caspase-6 down-regulation may more effectively suppress Lamin A/C cleavage indicating a more effective inhibition of caspase-6 30 activity; more effective inhibition of caspase-6 could lead to a greater stimulation of CNTF production, and better ROC survival and axon regeneration.
The involvement of caspases in axon regeneration The data presented here demonstrate that inhibition of caspase-6 along with down-regulation of caspase-2 promoted significant RGC axon regeneration and that this regeneration was approximately 10-fold greater than that observed using a pseudopeptide caspase inhibitor in previous studies (Monnier et aL, 2011). Notably, combined inhibition of caspase-2 and 5 caspase-6 significantly activated retinal glia. including Muller cells and astrocytes, and stimulated the production of high levels of CNTF both in;ivo and in vitro. Moreover, in combined caspase-2 and caspase-6 suppressed retinal cultures and in eyes, blocking CNTF receptor fhnction and JAK/STAT signalling both prevented glial activation, suppressed CNTF production and blocked RGC neurite/axon regeneration. These observations all 10 suggest a link between caspase-6 and retinal glia, Furthermore, inhibition of caspase-6 and caspase-2 promoted GFAP and CNTF expression in the glia present in the mixed retinal cultures, indicating an indirect mechanism of enhancing RGC survival and axon regeneration. Thus, without wishing to be bound by theory, the combination of caspase-2 and caspase-6 stimulates retinal gliosis, which in turn upregulates 15 the expression CNTF, and probably other neurotrophic factors, that then promote ROC survival and axon regeneration.
Furthermore, without being bound by theory, the current study implies differential mechanisms for RGC survival and axon regeneration and suggests that retinal glia are important to both processes. For example, the present inventors suggest that the activation of 20 retinal glia that occurs after ONC is enhanced by Penl-C6DN+siCASP2, increasing the release of CNTF. CNTF then acts not only on RGC in a paracrine manner to increase their regenerative capacity but also induces by an autocrine mechanism further reactive gliosis in astrocytes and Muller cells (DeChiara eta!, 1995, Kahn ci a!., 1995, Winter ci a!., 1995, Escartin et a!., 2006, Escartin et aL, 2007), which in turn enhances CNTF production, 25 contributing to an additional regenerative ability of RGC (Figure 9). CNTF is probably also released by RGC through autocrine mechanisms (Figure 9) contributing to the higher titres of CNTF in the combined Penl-C6DN+siCASIP2-treated retinae. This proposed mechanism might explain why enhanced RGC axon regeneration occurs afier Peni-C6DN+siCASP2 treatment. In contrast, blocking gpl3O or the JAK/STAT pathway does not prevent the low 30 levels of CNTF production by injury-induced gliosis but appears to suppress the C6DN-induced gliosis and subsequent high titre release of CNTF, which in turn reduces the regenerative response of RGC.
Mechanisms of caspase-6-modulated retinal gliosis and CNTF expression The present inventors observed that suppression of caspase-6 activates retinal microglia (Figure 0A-l0E) and thus may contribute to the release of pro-inflammatory cytokines.
Receptor binding of proinflammatory cytokines released from activated astrocytes and microglia activates a variety of intracellular signaling pathways, including the c-Jun N-5 terminal kinase (INK), p38 mitogen activated protein kinase (p38/MAPK), P13 kinase, extracellular signaling-related kinase (ERK) and activation of caspase-i and -3 (Van Eldik ci at, 2007, Anisman, 2009). Activation of JNK and p38/MAPK is known to contribute to the accumulation of GFAP in astrocytes (Tang ci at, 2006) and thus release from retinal glia of gpl3O receptor ligands (LIF and IL-6) which conspire with CNTF to signal RGC survival 10 through the JAK/STAT pathway. Indeed, CNTF is normally released by astrocytes and MUller cells of the retina after ONC, probably mediated by the release of inflammation-induced cytokines such as interleukin-1J3 and TNF-ct, all of which enhance CNTF release (Kamiguchi ci at, 1995, Muller ci at., 2007, Lorber ci at, 2008, Lorber dat, 2012), CNTF induces changes in astrocyte responses including upregulation of GFAP, cellular hypertrophy 15 and metabolic changes, that lead to the expression of further CNTF.
In conclusion, the present findings demonstrate that bi-therapies that suppress caspase-2 and caspase-6 inhibit ROC apoptosis and promote retinal gliosis, leading to the release of high titres of CNTF, which promotes RGC axon regeneration. These studies suggest that the collective targeting of caspase-2 and caspase-6 has therapeutic potential in treating human 20 adult nervous system trauma and disease.
Example 2: Animal Models for Testinft dsRNA Compounds iii Spinal Cord Injury In a non-limiting example, testing of the compositions disclosed herein comprising dsRNA inhibitors for treating spinal cord injury is performed in the rat spinal cord contusion model 25 as described by Young, 2002 (Young, 2002). Other predictive animal models of spinal cord injury are described in the following references: Gruner JA, 1992; 1-lasegawa, 2003; 1-luang and Young, 1994 (Gmner, 1992, 1-luang and Young, 1994, 1-lasegawa and Grumet, 2003).
Pharmaceutical compositions comprising a caspase-2 inhibitor and a caspase-6 inhibitor are tested in these animal models, which shows that these compositions treat spinal cord injury. 30 Example 3: Rat Models for Testing the caspase-2 inhibitor and the caspase-6 inhibitor in CNS Injury Closed Head Injury (CUD: Experimental traumatic brain injury (TBI) produces a series of events contributing to neurological and neurometabolic cascades, which are related to the degree and extent of behavioral deficits. CHI is induced under anesthesia, while a weight is 5 allowed to free-fall from a prefixed height (Chen et aL, 1996) over the exposed skull covering the left hemisphere in the midcoronal plane.
Transient middle cerebral artery occlusion (MCAO): A 90 to 120 minutes transient focal ischemia is performed in adult, male Sprague Dawley rats, 300-370 gr. The method employed is the intraluminal suture MCAO (Longa et at, 1989, Dogan et at, 1999). Briefly, under 10 halothane anesthesia, a 3-0-nylon suture material coated with Poly-L-Lysine is inserted into the right internal carotid artery (ICA) through a hole in the external carotid artery. The nylon thread is pushed into the ICA to the right MCA origin (20-23 mm). 90-t20 minutes later the thread is pulled off, the animal is closed and allowed to recover.
Permanent middle cerebral artery occlusion (MCAO): Occlusion is permanent, unilaterally-15 induced by electrocoagulation of MCA. Both methods lead to focal brain ischemia of the ipsilateral side of the brain cortex leaving the contralateral side intact (control). The left MCA is exposed via a temporal craniotomy, as described for rats (Tamura ci at, 1981). The MCA and its 20 lenticulostriatal branch are occluded proximally to the medial border of the olfactory tract with microbipolar coagulation. The wound is sutured, and animals returned to 20 their home cage in a room warmed at 26°C to 28°C. The temperature of the animals is maintained all the time with an automatic thermostat. Evaluation Process: The efficacy of the pharmaceutical compositions disclosed herein for treating CNS injury is determined by mortality rate, weight gain, infarct volume, short and long term clinical, neurophysiological and behavioral (including feeding behavior) outcomes in surviving animals. Infarct volumes 25 are assessed histologically (Mintorovitch c/at, 1991, Knight c/aL, 1994). The staircase test (Montoya ci at, 1991) or the motor disability scale according to Bederson's method (Bederson ci aT, 1986) is employed to evaluate the frmnctional outcome following MCAO.
The animals are followed for different time points, the longest one being two months. At each time point (24 hours, I week, 3, 6, 8 weeks), animals are sacrificed and cardiac perfusion 30 with 4% formaldehyde in PBS is performed. Brains are removed and serial coronal 200 jim sections are prepared for processing and paraffin embedding. The sections are stained with suitable dyes such as TCC, The infarct area is measured in these sections using a computerized image analyzer.
Pharmaceutical compositions comprising a RTP8OI inhibitor and a Casp2 inhibitor are tested in this animal model, which shows that these compositions treat and/or prevent CNS injury.
Example 4: APP Trans2enic Mouse Model of Alzheimer's Disease Animals and Treatment: The study includes twenty-four (24) APPV717I transgenic mice (female), a model for Alzheimer's disease (Moechars et aL, 1996, Moechars et at, 1999), aged 11 months that are randomly divided into two equal groups (Group I and Group II).
Animals are treated with a pharmaceutical composition comprising at least one siRNA 10 compound directed at the RTP8OI and/or Casp2 gene. Animals in control groups are treated with a vehicle solution. Compositions comprising the following concentrations of siRNA are tested: (i) 100 jig of siRNA compound/3 p1 of vehicle; (ii) 200 g of siRNA compound/3 ji 1 of vehicle and (Hi) 500 jig of siRNA compound / 3 p1 of vehicle, Compositions comprising the following vehicle are tested: (i) 5% glycerol solution; (ii) 10% glycerol solution and (iii) 15 15% glycerol solution, In this study the compositions are administered once every 4 days, during 3-4 month period of the experiment.
Termination: Mice are sacrificed; brains are dissected and processed as follows: one hemisphere for histological analysis and one hemisphere for molecular biology analysis.
Evaluation Process: The following histological analysis is performed: 20 1, Anti-amyloid 13 (A13) staining arid quantification (4 slides/mouse) 2, Thioflavin S staining and quantification of Af3 plaques(4 slides/mouse) 3. CD45 staining and quantification (4 slides/mouse) 4, GFAP (astrocytosis) staining and quantification Results: Pharmaceutical compositions comprising a caspase-2 inhibitor and a caspase-6 25 inhibitor are tested in this animal model, which shows that these composition are useful in treating Alzheimer's disease.
Example 5: Mouse Model of ALS Objective: To examine the efficacy of a caspase-2 inhibitor arid a caspase-6 inhibitor in the mutant SODI&9]A mouse model ofALS.
Animals and Treatment: Animals in test group are treated with at least one inhibitor of the caspase-2 gene and at least one inhibitor of the caspase-6 gene. Animals in control group are 5 treated with a composition comprising a control compound. Animals in vehicle group are treated with a vehicle solution. In this study the compositions are administered once every 4 days, starting from 30 days of age.
Analysis of disease progression: Behavioral and electromyography (EMG) analysis in treated and untreated mice is performed to monitor disease onset and progression. Mice are pre-10 tested before start of treatment, followed by weekly assessments, All results are compared statistically. The following tests are performed: 1. Swimming tank test: this test is particularly sensitive at detecting changes in hind-limb motor function (Raoul el at, 2005).
2. Electromyography: EMG assessments are performed in the gastrocnemius muscle of the 15 hind limbs, where compound muscle action potential (CMAP) is recorded (Raoul et cii., 2005).
3. Body weight: The body weight of mice is recorded weekly, as there is a significant reduction in the body weight of SODIG93A mice during disease progression (Kieran e at, 2007), Assessment of lifespan: The lifespan in days for treated and untreated mice is recorded 20 and compared statistically to determine whether treatment by administering a caspase-2 inhibitor and a caspase-6 inhibitor implicated in ALS has any significant effect on lifespan.
Mice are sacrificed at a welidefined disease end point, when they have lost >20% of body weight and are unable to raise themselves in under 20 seconds. All results are compared statistically. 25 Post mortem histopathology: At the disease end-point mice are terminally anaesthetized and spinal cord and hind-limb muscle tissue are collected for histological and biochemical analysis.
Examining motor neuron survival: Transverse sections of lumbar spinal cord are cut using a cryostat and stained with gallocyanin, a nissl stain. From these sections the number of motor 30 neurons in the lumbar spinal cord is counted (Kieran el at., 2007), to determine whether dsRNA treatment prevents motor neuron degeneration in SOD1G93A mice.
Examining spinal cord histopathology: Motor neuron degeneration in SOD]093A mice results in astrogliosis and activation of microglial cells, Here, using transverse sections of lumbar spinal cord the activation of astrocytes and microglial cells is examined using 5 immunocytochemistry to determine whether combined caspase-2 inhibitor and caspase-6 inhibitor treatment reduced or prevented their activation.
Examining muscle histology. Hind-limb muscle denervation and atrophy occur as a consequence of motor neuron degeneration in SOD 1093A mice, At the disease end point the weight of individual hind-limb muscles (gastrocnemius, soleus, tibialis anterior, extensor Jo digitorium longus muscles) is recorded and compared between treated and untreated mice.
Muscles are then processed histologically to examine motor end plate denervation and muscle atrophy (Kieran c/aL, 2005).
Example 6: Model Systems of Huntin2ton's Disease (HP) in Mice 15 In a non-limiting example, testing of the compositions disclosed herein for treating Huntington's disease is performed in the HD mouse model, R6/2 (Wang c/at, 2005).
Results: The compositions and methods disclosed herein are tested in this animal model, and show efficacy in treating Huntington's disease, Example 7: Evaluation of Protection of Retinal Gan2lion Cell Dendrites after Axonal Injury in RGC-YFP Trans2enic Mice Model Experimental animals and surgical procedure: Experimental procedures are carried out on CS7BL/6 transgenic or wild-type control mice. Adult transgenic mice carrying the yellow fluorescent protein (YFP) gene under control of the Thy-i promoter (YFP-H line, Jackson 25 Laboratory, Bar Harbor, ME, USA;) are studied (Feng c/at., 2000). Approximately 10-30% of retinal ganglion cells are exclusively labelled in the retina of these transgenic mice. All surgical procedures were carried out on 3 to 7 month-old mice under general anesthesia (2% Isoflurane; 0.8 L/min).
Optic nerve axotomy: The optic nerve axotomy is carried out on mice as previously described (Lebmn-Julien et at, 2009). Briefly, the left optic nerve iss exposed and carefully transected at 05-1 mm from the optic nerve head. During this procedure care is taken to avoid injury to the ophthalmic artery. Fundus examination is routinely performed immediately after axotomy and 3 days later to verify the integrity of the retinal circulation after surgery. Mimals 5 showing signs of compromised blood supply are excluded from the study.
Intravitreal injection: caspase-2 and caspase-6 inhibitors are used in this study. The inhibitors are administered by a single intravitreal injection (2 pI) into the vitreous chamber of the left eye of YFP mice at the time of the optic nerve injury. The intravitreal inj ections were made using a 10 R1 Hamilton syringe adapted with a 32 gauge glass micro needle as described 10 previously (Lebrun-Julien et at, 2009). Briefly, the micro needle is introduced in the superior hemisphere of the ocular globe. During this procedure care is taken to avoiding lens injury by introducing the micro needle at an angle of 45 degree through the sclera. The injection is performed over a period of 2 minutes and the needle is held still during another 2 minutes to enable the inhibitors to diffuse into the vitreous chamber, After the injection, surgical glue is (Indermill, Tyco Health Care, Mansfield, MA, USA) is immediately used to seal the site of injection, avoiding any leakage.
Results: The compositions disclosed herein are tested in this animal model, and show efficacy in protection of retinal ganglion cell dendrites after axonal injury.
Example 8: Oxygen-Induced Retinopathy (OTR1 Rat Model for Evaluation of Protection of Retinal Ganglion Cells following Ischemia-Reperfusion Injury Oxygen-Induced Retinopathy (OIR) model is a relevant model for angle closure glaucoma.
Methods Animal Model: Male Brown Norway rats (Japan SLC, inc., Shizuoka) weighing 25 approximately 200 to 250 g each are used in accordance with the ARVO Statement for the Use of Animals in Vision and Ophthalmic Research. Only one eye of each rat is used, The rats are anesthetized for all procedures with a mixture (1:1) of xylazine hydrochloride (4 mg/kg) (Bayer, Tokyo, Japan) and ketamine hydrochloride (10 mg/kg) (Sankyo, Tokyo, Japan), and the ocular surface is then anesthetized with topical instillation of 0.4% 30 oxybuprocaine hydrochloride (Santen, Osaka, Japan), The pupils are dilated with 0,5% tropicamide and 2.5% phenylephrine hydrochloride (Santen).
lschemia-Reperfusion: The rats are placed under deep anesthesia with intramuscular injection of ketamine and xylazine. Ischemia is applied to the eye by increasing the intraocular pressure to cut off the blood supply from the retinal artery. Increased pressure is achieved by introduction of sterile saline through a 30-gauge needle that is inserted into the anterior chamber of the eye through the cornea. Each anterior chamber is cannulated with a 30-gauge 5 inifision needle connected to a normal saline (0.9% sodium chloride) container through tubing (TI-U450P07, Terumo, Tokyo). The TOP in the cannulated eyes is raised to 90 mmHg for a period of 90 mm by elevating the saline container. Intraocular pressures is measured using a rebound microtonometer designed for use on rodent eyes (TonoLab, bare, Helsinki, Finland). Total eye ischemia is evident from the whitening of the anterior segment of the eye 10 and the blanching of the retinal arteries on fundus examination. At the end of the ischemic period, the needle is removed from the anterior chamber, and reperfusion of the retinal vasculature is confirmed, FG labeling: Rats are anaesthetized and sterile eye lubricant ointment is applied to prevent drying of the corneas during surgery. Head fur is shaved (from eye to ear level) and the head 15 is fixed on the head stage by a head clamp. Operation area is disinfected with 10% povidone iodine solution followed by 70% alcohol, The point of Fluor-Gold injection is designated at a depth of 3,5 mm from the brain surface, 6.5 mm behind the bregma, 2.0 mm lateral to the midline. A hole is drilled in the skull and at the superior colliculi are injected with 2.5 micro litter of 4% FG (Fluorochrome, Inc. 529400, Englewood, CO). 20 RGC counts: Labeled RGCs are counted in photographs taken from 12 areas (0,2 x 0,2 mm) of each retina situated, three in every retinal quadrant from the optic disc. The number of labeled cells in the photographs is divided by the area of the region to obtain mean densities of labeled cells per square millimeter, and the densities obtained in the 12 areas are pooled to calculate a mean RGC density per retina. Distinguishable glial cells (bright and small cells) 25 are not counted. Cell counts are performed in a masked fashion.
Optical Coherence I'omography (OCI') retinal thickness analysis: OCt is based on low coherence interferometrv and provides high-resolution cross-sectional images of the retina.
Retinal thickness was measured with OCT (Cirrus OCt Carl Zeiss Meditec, Inc., Dublin, CA), After maximal pupillary dilatation, anesthetized rats are mounted in a head holder. The 30 optic disc is placed in the center of the OCT image, and the scanning line is aligned to pass through both the inner and outer canthi. The scan length was 5.0 25 mm in all cases. Retinal thickness is measured by OCT at 1-disc diameters from the optic disc margin in the peripheral retina with an accessory program of the OCT instrument. The mean retinal thickness of one eye is defined as the average of the three measurements. All image analyses are performed in a masked fashion.
Results: The compositions disclosed herein are tested in this animal model, and show efficacy in preservation of retinal thickness. 5 Example 9: Rat Axotomy Model for Evaluation of the Neuroprotective Effect of a combination of a Caspase-2 inhibitor and a Caspase-6 inhibitor Evaluation of the neuroprotective effect of a combination of a caspase-2 inhibitor and a caspase-6 inhibitor after administration by intravitreal (IVT) is performed in Rat Axotomy 10 Model at two (2) weeks post injury.
Objective: to evaluate potential additive or synergistic effects of a caspase-2 inhibitor and a caspase-6 inhibitor in RGC neuroprotection in optic nerve axotomy model.
Methodology: Optic nerve axotomy is performed in adult rats, Immediately after surgery, rats receive intravitreal injections (injection volume is S uL) with a negative control or with the a 15 combination of a caspase-2 inhibitor and a caspase-6 inhibitor. Second similar injections are performed into corresponding eyes at 1 week after axotomy. Evaluation of the neuroprotective effects of each of the treatments is performed by counting of PG relabeled RGC in retinal whole mounts at 2 weeks after axotomy.
Results: The compositions disclosed herein are tested in this animal model, and show efficacy 20 in providing neuroprotection.
REFERENCES
Agudo NI, Perez-Mann MC, Lonngren U, Sobrado P, Conesa A, Canovas I, eta!. Time course profiling of the retinal transcriptome after optic nerve transection and optic nerve cmsh. Mo!ecular vision. 2008; 14:1050-63 Ahmed Z, Aslam M, Lorber B, Suggate EL, Berry M, Logan A. Optic nerve and vitrea! 5 inflammation are both RGC neuroprotective but on!y the!atter is RGC axogenic.
Neurobiology of disease. 201 0;37:44]-54.
Ahmed Z, Dent RG, Leadbeater WE, Smith C, Berry M, Logan A. Matrix meta!!oproteases: degradation of the inhibitory environment of the transected optic nerve and the scar by 10 regenerating axons, Molecular and cellular neurosciences. 2005;28:64-78.
Ahmed Z, Kalinski H, Berry NI, Almasieh Tvl, Ashush H, Slager N, eta!. Ocu!ar neuroprotection by siRNA targeting caspase-2. Cel! death & disease. 201 1;2:e173.
Ahmed Z, Suggate EL, Brown ER, Dent RG, Armstrong SJ, Barrett LB, et al. Schwann ccl!-derived factor-induced modulation of the NgR/p75NTR!EGFR axis disinhibits axon growth through CNS myelin in vivo and in vitro, Brain, 2006; 129:1517-33, Akpan N, Serrano-Saiz E, Zacharia BE, Otten ML, Ducruet AF, Snipas SJ, et al. Intranasal 20 de!ivery of caspase-9 inhibitor reduces caspase-6-dependent axon/neuron!oss and improves neuro!ogica! function after stroke. Journa! of neuroscience. 2011;3 1:8894-904.
Anisman H. Cascading effects of stressors and inflammatory immune system activation: imp!ications for major depressive disorder. Journa! of psychiatry & neuroscience. 2009;34:4-25 20.
Bederson JB, Pifts LH, Tsuji M, Nishimura MC, Davis RL, Bartkowski H, Rat midd!e cerebral artery occlusion: evaluation of the model and development of a neurologic examination, Stroke. 1986; 17:472-6, 30 Berkelaar NI, C!arke DB, Wang YC, Bray GM. Aguayo AJ. Axotomy resu!ts in de!ayed death and apoptosis of retina! gang!ion ce!ls in adu!t rats, Journal of neuroscience.
1994; 14:4368-74.
Berry NI, Car!i!e J, Hunter A. Periphera! nerve exp!ants grafted into the vitreous body of the eye promote the regeneration of retina! gang!ion ce!! axons severed in the optic nerve, Journal of neurocytology. 1996;25: 147-70.
Berry NI, Carlile J, Hunter A, Tsang W, Rosenstiel P, Sievers J. Optic nerve regeneration 40 after intravitreal periphera! nerve implants: trajectories of axons regrowing through the optic chiasm into the optic tracts. Journa! of neurocyto!ogy. 1 999;28:721-41.
Bo!wch GM, Caspase-mediated activation of PAK2 during apoptosis: proteo!ytic kinase activation as a general mechanism of apoptotic signal transduction? Cell death and 45 differentiation. 1998;5:637-45.
Casciola-Rosen LA, Mi!!er DK, Anha!t GJ, Rosen A. Specific cleavage of the 70-kDa protein component of the Ui sma!l nuc!ear ribonucleoprotein is a characteristic biochemical feature of apoptotic ce!l death. The Journa! of bio!ogical chemistry, 1994;269:30757-60.
Chen Y, Constantini S, Trembovier V, Weinstock M, Shohami B. An experimental model of dosed head injury in mice: pathophysiology, histopathology, and cognitive deficits. Journal of Neurotrauma. 1996; 13: 557-68.
Davidson TJ, 1-lard 5, Arboleda VA, Prunell GF, Shelanski ML, Greene LA, et al, Highly efficient small interfering RNA delivery to primary mammalian neurons induces MicroRNA-like effects before mRNA degradation. Journal of Neuroscience, 2004;24: 10040-6.
DeChiara TM, Vejsada R, Poueymirou WI, Acheson A, Sun C, Conover JC, et al. Mice 10 lacking the CNTF receptor, unlike mice lacking CNTF, exhibit profound motor neuron deficits at birth. Cell. l995;83:313-22.
Dogan A, Rao AM, Hatcher I, Rao VL, Baskaya MX, Dempsey RJ. Effects of MDL 72527, a specific inhibitor of polyamine oxidase, on brain edema, ischemic injury v&ume, and tissue 15 polyamine levels in rats after temporary middle cerebral artery occlusion, Journal of Neurochemistry. 1999;72:765-70.
Douglas MR, Morrison KC, Jacques Si, Leadbeater WE, Gonzalez AM, Berry M, et al, Off-target effects of epidermal growth factor receptor antagonists mediate retinal ganglion cell 20 disinhibited axon growth. Brain. 2009;132:3102-21.
Edgington LE, van Raam BJ, Verdoes NI, Wierschem C, Salvesen GS, Bogyo M. An optimized activity-based probe for the study of caspase-6 activation, Chemistry & Biology, 2012;19:340-52. 25 Bscartin C, Brouillet B, Gubellini P, Trioulier Y, Jacquard C, Smadja C, et al. Ciliary neurotrophic factor activates astrocytes, redistributes their glutamate transporters GLAST and GLT-t to raft microdomains, and improves glutamate handling in vivo, Journal of Neuroscience 2006;26:5978-89, 30 Bscartin C, Pierre K, Cohn A, Brouillet B, Delzescaux T, Guillermier lvi, et al. Activation of astrocytes by CNTF induces metabolic p'asticity and increases resistance to metabolic insults.
Journal of Neuroscience, 2007;27:7094-104, Feng G, Mellor RH, Bernstein M, Keller-Peck C, Nguyen QT, Wallace lvi, et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron.
2000;28:4t-51, Garcia-Valenzuela F, Gorczyca W, Darzynkiewicz Z, Sharma SC, Apoptosis in adult retinal 40 ganglion cells after axotomy. iournal of Neurobiology. 1994;25:43]-8.
Gruner JA. A monitored contusion model of spinal cord injury in the rat. Journal of Neurotrauma, I 992;9: t23-6; discussion 6-8.
Hasegawa K, Grumet Ni. Trauma-induced tumorigenesis of cells implanted into the rat spinal cord. iournal of Neurosurgery. 2003;98: 065-71.
Huang PP, Young W. The effects of arterial blood gas values on lesion volumes in a graded rat spinal cord contusion model. Journal of Neurotrauma, 1994; 11:547-62, 50 Isenmann 5, WahI C, Krajewski 5, Reed SC, Bahr M. Up-regulation of Bax protein in degenerating retinal ganglion cells precedes apoptotic cell death after optic nerve lesion in the rat. European Journal of Neuroscience. 1997;9:1763-72.
Kahn MA, Ellison JA, Speight GJ, de Vellis J, CNTF regulation of astrogliosis and the activation of microglia in the developing rat central nervous system. Brain Research.
I 995;685:55-67.
Kamiguchi H, Yoshida K, Sagoh M, Sasaki H, Inaba M, Wakamoto H, et al. Release of 10 ciliary neurotrophic factor from cultured astrocytes and its modulation by cytokines.
Neurochemical Research. I 995;20:1187-93 Kermer P, Klocker N, Labes M, Bahr M. Inhibition of CPP32-like proteases rescues axotomized retinal ganglion cells from secondary cell death in vivo. Journal of Neuroscience. 15 1998;18:4656-62.
Kieran D, Hafezparast M, Bohnert S, Dick JR, Martin J, Schiavo G, et al. A mutation in dynein rescues axonal transport defects and extends the life span of ALS mice. Journal of Cell Biology. 2005;169:561-7. 20 Kieran D, Woods I, Villunger A, Strasser A, Prehn JH. Deletion of the BH3-only protein puma protects motoneurons from ER stress-induced apoptosis and delays motoneuron loss in ALS mice. Proceedings of the National Academy of Sciences USA. 2007; 104:20606-11.
Kirsch lvi, Trautmann N, Ernst M, Hofmann HD. involvement of gpl3O-associated cytokine signaling in Muller cell activation following optic nerve lesion. Glia. 2010;58:768-79.
Knight RA, Dereski MO, Helpern JA, Ordidge RJ, Chopp M. Magnetic resonance imaging assessment of evolving focal cerebral ischemia. Comparison with histopathology in rats. 30 Stroke, I 994;25: 1252-61; discussion 61-2.
Lazebnik YA, Kaufmann SH, Desnoyers 5, Poirier GO, Earnshaw WC. Cleavage of poly(ADP-ribose) polymerase by a proteinase with properties like ICE. Nature.
1994;371:346-7. 35 Lebmn-Julien F, Duplan L, Pemet V, Osswald I, Sapieha P, Bourgeois P, et al. Excitotoxic death of retinal neurons in vivo occurs via a non-cell-autonomous mechanism, Journal of Neuroscience 2009;29:5536-45, Leist M, Single B, Castoldi AF, Kuhnle 5, Nicotera P. Intracellular adenosine triphosphate (AlP) concentration: a switch in the decision between apoptosis and necrosis. Journal of Experimental Medicine. 1997; 185:1481-6.
Leon 5, Yin Y, Nguyen J, Irwin N, Benowitz LI, Lens injury stimulates axon regeneration in 45 the mature rat optic nerve. Journal of Neuroscience, 2000;20:4615-26.
Longa EZ, Weinstein PR, Carlson 5, Cummins R. Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke. 1989;20:84-91.
Lorber B, Berry Nil, Douglas MR, Nakazawa T, Logan A. Activated retinal glia promote neurite outgrowth of retinal ganglion cells via apolipoprotein B. Journal of Neuroscience Research. 2009;87:2645-52.
Lorber B, Berry M, Logan A. Lens injury stimulates adult mouse retinal ganglion cell axon 5 regeneration via both macrophage-and lens-derived factors. European Journal of Neuroscience. 2005;2 t:2029-34, Lorber B, Berry Nil, Logan A. Different factors promote axonal regeneration of adult rat retinal ganglion cells after lens injury and intravitreal peripheral nerve grafting. Journal of 10 Neuroscience Research, 2008;86:894-903.
Lorber B, Berry Nil, Logan A, Tonge D. Effect of lens lesion on neurite outgrowth of retinal ganglion cells in vitro. Molecular and Cellular Neuroscience. 2002;21:301-11.
Lorber B, Guidi A, Fawcett JW, Martin KR, Activated retinal glia mediated axon regeneration in experimental glaucoma. Neurobiology of Disease. 201 2;45:243-52.
Mintorovitch J, Moseley N'IE, Chileuitt L, Shimizu H, Cohen Y, Weinstein PR. Comparison of diffusion-and T2-weighted v1IRI for the early detection of cerebral ischemia and 20 reperfusion in rats. Magnetic Resonance in Medicine. 1991;18:39-50.
Mintzer R, Ramaswamy S, Shah K, Hannoush RN, Pozniak CD, Cohen F, et al. A whole cell assay to measure caspase-6 activity by detecting cleavage of lamin A/C, PbS One.
2012;7:e30376. 25 Moechars D, Lorent K, De Strooper B, Dewachter I, Van Leuven F. Expression in brain of amyboid precursor protein mutated in the alpha-secretase site causes disturbed behavior, neuronal degeneration and premature death in transgenic mice, The EMBO journal.
1996;15:1265-74. 30 Moechars D, Lorent K, Van Leuven F. Premature death in transgenic mice that overexpress a mutant amyloid precursor protein is preceded by severe neurodegeneration and apoptosis.
Neuroscience. t999;9t:819-30, Monnier PP, D'Onofrio PM, Magharious M, Hollander AC, Tassew N, Szydlowska K, et al. Involvement of caspase-6 and caspase-8 in neuronal apoptosis and the regenerative failure of injured retinal ganglion cells, Journal ofNeuroscience. 20 t t 3 1:10494-505, Montoya CP, Campbell-Hope U, Pemberton KD, Dunnett SB. The "staircase test": a 40 measure of independent forelimb reaching and grasping abilities in rats. Journal of Neuroscience Methods. 1991;36:2 t9-28.
Muller A, Hauk TG, Fischer D, Astrocyte-derived CNTF switches mature RGCs to a regenerative state following inflammatory stimulation. Brain, 2007; 130:3308-20, 45 Nikolaev A, McLaughlin T, O'Leary DD, Tessier-Lavigne Nil. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature. 2009;457:981-9.
Orth K, Chinnaiyan AM, Garg M, Froelich Ci, Dixit VI'vl. The CED-3/ICE-like protease Mch2 is activated during apoptosis and cleaves the death substrate lamin A. Journal of Biological Chemistry. 1 996;271:16443-6.
Peinado-Ramon P, Salvador M, Villegas-Perez MP, Vidal-Sanz M. Effects of axotomy and 5 intraocular administration ofNT-4, NT-3, and brain-derived neurotrophic factor on the survival of adult rat retinal ganglion cells. A quantitative in vivo study. Investigative Ophthalmology & Visual Science. I 996;37:489-500.
Pernet V, Di Polo A. Synergistic action of brain-derived neurotrophic factor and lens injury 10 promotes retinal ganglion cell survival, but leads to optic nerve dystrophy in vivo. Brain.
2006;129:1014-26.
Pop C, Salvesen GS. Human caspases: activation, specificity, and regulation. Journal of Biological Chemistry. 2009;284:2 1777-81. 15 Porter AG, Ng P, Janicke RU. Death substrates come alive. BioEssays, 1997;19:501-7, Rabacchi SA, Bonfanti L, Liu XI-l, Mafl'ei L. Apoptotic cell death induced by optic nerve lesion in the neonatal rat. Journal of Neuroscience. 1994; 14:5292-301. 20 Raoul C, Abbas-Terki T, Bensadoun JC, Guillot 5, Haase U, Szulc J, et al. Lentiviral-mediated silencing of SOD! through RNA interference retards disease onset and progression in a mouse model of ALS, Nature Medicine. 2005;ii:423-8.
Ruchaud 5, Korfali N, Villa P, Kottke Ti, Dingwall C, Kaufmann SH, et al. Caspase-6 gene disruption reveals a requirement for lamin A cleavage in apoptotic chromatin condensation.
The EMBO journal. 2002;2 I: 1967-77.
Song Q, Lees-Miller SP, Kumar 5, Zhang Z, Chan DW, Smith GC, et al, DNA-dependent 30 protein kinase catalytic subunit: a target for an ICE-like protease in apoptosis. The EMBO journal. 1996;15:3238-46.
Takahashi A, Alnemri ES, Lazebnik YA, Fernandes-Alnemri T, Litwack G, Moir RD, et al, Cleavage of lamin A by Mch2 alpha but not CPP32: multiple interleukin 1 beta-converting 35 enzyme-related proteases with distinct substrate recognition properties are active in apoptosis. Proceedings of the National Academy of Sciences USA. 1996;93:8395-400.
Tamura A, Graham DI, McCulloch J, Teasdale GM. Focal cerebral ischaemia in the rat: 1.
Description of technique and early neuropathological consequences following middle 40 cerebral artery occlusion. iournal of Cerebral Blood Flow and Metabolism. 198] ;1:53-60.
Tang G, Xu Z, Goldman JE. Synergistic effects of the SAPK/JNK and the proteasome pathway on glial fibrillary acidic protein (GFAP) accumulation in Alexander disease. Journal of Biological Chemistry. 2006;28 1:38634-43. 45 Thanos 5, Vidal-Sanz Ni, Aguayo AJ. The use of rhodamine-B-isothiocyanate (RITC) as an anterograde and retrograde tracer in the adult rat visual system. Brain Research.
1987;406:317-21.
Van Eldik U, Thompson WL, Ralay Ranaivo H, Behanna HA, Martin Watterson D. Glia proinflammatory cytokine upregulation as a therapeutic target for neurodegenerative diseases: function-based and target-based discovery approaches. International Review of Neurobiology. 2007;82:277-96.
Vigneswara V. Berry M, Logan A, Ahmed Z. Pharmacological inhibition of caspase-2 protects axotomised retinal ganglion cells from apoptosis in adult rats. PbS One.
2012;7:e53473.
Villegas-Perez MP, Vidal-Sanz M, Rasminsky lvi, Bray GM, Aguayo AJ. Rapid and 10 protracted phases of retinal ganglion cell loss follow axotomy in the optic nerve of adult rats.
Journal of Neurobiology. I 993;24:23-36.
Wang YL, Liu W, Wada B, Murata M, Wada K, Kanazawa I. Clinico-pathological rescue of a model mouse of Huntington's disease by siRNA. Neuroscience Research. 2005;53:241-9. 15 Winter CG, Saotome Y, Levison SW, Hirsh D. A role for ciliary neurotrophic factor as an inducer of reactive gliosis, the glial response to central nervous system injury. Proceedings of the National Academy of Sciences USA. 995;92:5865-9.
Yin Y, Cui Q, Li Y, Irwin N, Fischer D, Harvey AR, et al. Macrophage-derived factors stimulate optic nerve regeneration. Journal of Neuroscience. 2003;23:2284-93.
Young W. Spinal cord contusion models. Progress in Brain Research, 2002; 137:231-55.

Claims (7)

  1. CLAIMS: 1 An inhibitor of caspase-2 for use in the treatment of a disease, a disorder or an injury of the nervous system by combined, sequential or separate administration with an inhibitor of caspase-6.
  2. 2. An inhibitor of caspase-2 for use according to claim 1, wherein the nervous system is selected from the group consisting of the central nervous system (CNS) and the peripheral nervous system (PNS).
  3. 3, An inhibitor of caspase-2 for use according to claim 2, wherein the nervous system comprises the central nervous system (CNS).
  4. 4, An inhibitor of caspase-2 for use according to claim 3, wherein the central nervous system (CNS) comprises the visual system and the audio-vestibular system.
  5. 5. An inhibitor of caspase-2 for use according to any one of claims 3 to 4, wherein the disease, disorder or injury is selected from the group consisting of an ocular disease; an ocular disorder or an ocular injury; a spinal cord disease, a spinal cord disorder or a spinal cord injury; a brain disease, a brain disorder or a brain injury; and a disease, disorder or injury of the audio-vestibular system.
  6. 6. An inhibitor of caspase-2 for use according to any one of claims 3 to 5, wherein the disease, disorder or injury is selected from the group consisting of a neurodegenerative disease and a neurological disorder,
  7. 7. An inhibitor of caspase-2 for use according to any one of claims 3 to 6, wherein the disease, disorder or injury is selected from the group consisting of inflammation, neurotoxicity, oxidative stress, and traumatic damage due to a tumor growth, or a combination thereof 8 An inhibitor of caspase-2 for use according to claim 6, wherein the neurodegenerative disease is selected from the group consisting of an acute neurodegenerative disease and a chronic neurodegenerative disease.9. An inhibitor of caspase-2 for use according to claim 3 to 4, wherein the disease, disorder or injury of the central nervous system (CNS) is an ocular disease, an ocular disorder or an ocular injury.ID. An inhibitor of caspase-2 for use according to claim 9, wherein the ocular disease, ocular disorder or ocular injury is selected from the group consisting of neurodegeneration, inflammation, and oxidative stress, or a combination thereof II An inhibitor of caspase-2 for use according to claim 9, wherein the ocular disease, ocular disorder or ocular injury is selected from the group consisting of glaucoma, and diabetic retinopathy (DR).12. An inhibitor of caspase-2 for use according to claim 9, wherein the ocular disease, ocular disorder or ocular injury is an optic neuropathy selected from the group consisting of ischemic optic neuropathy (ION), hereditary optic neuropathy, metabolic optic neuropathy, neuropathy due to a toxic agent, neuropathy caused by adverse drug reactions and neuropathy caused by vitamin deficiency.13. An inhibitor of caspase-2 for use according to claim 12, wherein the ischemic optic neuropathy comprises non-arteritic ischemic optic neuropathy (NAION).14. An inhibitor of caspase-2 for use according to claim 12, wherein the hereditary optic neuropathy comprises Leber' s hereditary optic neuropathy (LEJON), 15. An inhibitor of caspase-2 for use according to claim 9, wherein the ocular disease, ocular disorder or ocular injury is selected from the group consisting of optic neuritis, retinal artery occlusion, central retinal vein occlusion, brunch retinal vein occlusion and optic nerve injury.16. An inhibitor of caspase-2 for use according to claim 9, wherein the ocular disease, ocular disorder or ocular injury is selected from the group consisting of ischemic injury, ischemia-reperfusion injury, mechanical injury, injury or interruption of nerve fibers, physically damaged nerve, neurite damage, and a condition associated with lack of retrograde supply of neurotrophic factor.17. An inhibitor of caspase-2 for use according to any one of claims 1 to 4, wherein the treatment provides neuroprotection.18. An inhibitor of caspase-2 for use according to any one of claims to 4 or 17, wherein the treatment promotes axon regeneration of neuronal cells.19. An inhibitor of caspase-2 for use according to claim 18, wherein the neuronal cells are cells of the peripheral nervous system (PNS).20. An inhibitor of caspase-2 for use according to claim 18, wherein the neuronal cells are cells of the central nervous system (CNS).2]. An inhibitor of caspase-2 for use according to claim L/, wherein the neuroprotection comprises ocular neuroprotection.22. An inhibitor of caspase-2 for use according to claim 17, wherein the neuroprotection comprises audio-vestibular neuroprotection.23. An inhibitor of caspase-2 for use according to claim 5, wherein the treatment promotes axon regeneration of retinal neuronal cells or dorsal root ganglion cells.24. An inhibitor of caspase-2 for use according to any one of claims 17 to 22, wherein the neuroprotection is selected from the group consisting of protection of neural cells from apoptosis, promoting survival of neural cells, increasing the number of neural cell neuritis, increasing neurite cell outgrowth, promoting retinal gliosis, promoting regeneration of neural cells and increasing or stimulation of neurotrophic factors in the nervous system.25, An inhibitor of caspase-2 for use according to claim 24, wherein the neural cell comprises a ganglion cell.26. An inhibitor of caspase-2 for use according to claim 25, wherein the ganglion cell is selected from the group consisting of a retinal ganglion cell, a spiral ganglion cell, a vestibular ganglion cell, a dorsal ganglion cell and a peripheral ganglion cell.27. An inhibitor of caspase-2 for use according to claim 24, wherein the neural cell comprises a retinal neural cell.28. An inhibitor of caspase-2 for use according to claim 27, wherein the retinal neural cell is selected from the group consisting of a photoreceptor cell, a bipolar cell, a ganglion cell, a horizontal cell and an amacrine cell, 29. An inhibitor of caspase-2 for use according to claim 28, wherein the retinal neural cell comprises a retinal ganglion cell (ROC).30. An inhibitor of caspase-2 for use according to any one of claims t 7 to 24, wherein the neuroprotection comprises protecting neurons from death, 31. An inhibitor of caspase-2 for use according to claim 30, wherein death of the neuron is associated with one or more of a disease or disorder, a surgery, ischemia, ischemia/reperfusion, physical/mechanical trauma, a chemical agent, an infectious agent, an immunologic reaction and a nutritional imbalance.32. An inhibitor of caspase-2 for use according to any one of claims 30 to 3], wherein the inhibitor of caspase-2 and the inhibitor of caspase-6 are independently configured for contacting the neuron.33, An inhibitor of caspase-2 for use in providing neuroprotection and axon regeneration of neuronal cells by combined, sequential or separate administration with an inhibitor of caspase-6.34. An inhibitor of caspase-2 for use according to any one of claims] to 33, wherein the inhibitor of caspase-2 and the inhibitor of caspase-6 are independently selected from the group consisting of a small organic molecule, a protein, an antibody or fragment thereof, a peptide, a polypeptide, a peptidomimetic and a nucleic acid molecule; or a salt or prodrug thereof 35. An inhibitor of caspase-2 for use according to claim 34, wherein at least one of the inhibitor of caspase-2 and the inhibitor of caspase-6 is a nucleic acid molecule independently selected from the group consisting of a single stranded antisense nucleic acid (ssNA), a double-stranded NA (dsNA), a small interfering NA (siNA), a short hairpin NA (shNA), a micro RNA (miRNA), an aptamer, and a ribozyme, or a salt or prodrug thereof 36. An inhibitor of caspase-2 for use according to claim 35, wherein each of the ssNA or the dsNA independently comprises one or more of a modified nucleotide, an unmodified nucleotide, a nucleotide analogue and an unconventional moiety.37. An inhibitor of caspase-2 for use according to claim 36, wherein at least one of the inhibitor of caspase-2 and the inhibitor of caspase-6 is a double-stranded NA (dsNA) independently selected from an unmodified double-stranded NA (dsNA) or a chemically modified double-stranded NA (dsNA; or a salt or prodrug thereof 38. An inhibitor of caspase-2 for use according to claim 37, wherein the inhibitor of caspase-2 compiises an antisense strand sequence 5' AGGAGUTJCCACAUUCUGGC 3' and a sense strand sequenceS' GCCAGAAUGUGGAACUCCTJ 3'.39. An inhibitor of caspase-2 for use according to claim 38, wherein the antisense strand of the inhibitor of caspase-2 comprises 2'-O-methyl sugar modified ribonucleotides in positions (5>3') 2, 4, 6, 8, 11, 13, 15, 17 and 19; and wherein the sense strand of the inhibitor of caspase-2 comprises an [-DNA (L-deoxycytidine) nucleotide in position 18 and an inverted deoxyabasic moiety covalently attached at the 5' terminal of the strand.40. An inhibitor of caspase-2 for use according to any of claims I to 39, for concurrent administration with the inhibitor of caspase-6.41. An inhibitor of caspase-2 for use according to claim 40, wherein the inhibitor of 5 caspase-2 and the inhibitor of caspase-6 are administered in a single dosage form, 42. An inhibitor of caspase-2 for use according to claim 41, wherein the inhibitor of caspase-2 and the inhibitor of caspase-6 are administered in separate dosage forms, 43. An inhibitor of caspase-2 for use according to any of claims 1 to 39, for sequential administration with the inhibitor of caspase-6. 10 44. The inhibitor of caspase-2 for use according to claim 41, wherein the inhibitor of caspase-2 and the inhibitor of caspase-6 comprise different portions of a single molecule.45. An inhibitor of caspase-2 for use according to any of claims to 39, for combined, sequential or separate administration with a neurotrophic factor.46. An inhibitor of caspase-2 for use according to claim 45, wherein the neurotrophic factor is selected from the group consisting of ciliary neurotrophic factor (CNTF), brain-derived neurotrophic factor, nerve growth factor, neurotrophin-3 and fibroblast growth factor-is 2, or any combination thereof 47. A composition comprising an inhibitor of caspase-2 and an inhibitor of caspase-6.
GB1404289.9A 2014-03-11 2014-03-11 Combined inhibition of caspase-2 and caspase-6 in neuroprotection and axon regeneration Withdrawn GB2526767A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
GB1404289.9A GB2526767A (en) 2014-03-11 2014-03-11 Combined inhibition of caspase-2 and caspase-6 in neuroprotection and axon regeneration

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB1404289.9A GB2526767A (en) 2014-03-11 2014-03-11 Combined inhibition of caspase-2 and caspase-6 in neuroprotection and axon regeneration

Publications (2)

Publication Number Publication Date
GB201404289D0 GB201404289D0 (en) 2014-04-23
GB2526767A true GB2526767A (en) 2015-12-09

Family

ID=50554886

Family Applications (1)

Application Number Title Priority Date Filing Date
GB1404289.9A Withdrawn GB2526767A (en) 2014-03-11 2014-03-11 Combined inhibition of caspase-2 and caspase-6 in neuroprotection and axon regeneration

Country Status (1)

Country Link
GB (1) GB2526767A (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117427152B (en) * 2023-12-20 2024-05-24 中国人民解放军军事科学院军事医学研究院 Application of caspase-2 to removal of pathological overload ubiquitin aggregate

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006056487A2 (en) * 2004-11-24 2006-06-01 Theraptosis S.A. Peptides useful as dual caspase-2/-6 inhibitors and their biological applications

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006056487A2 (en) * 2004-11-24 2006-06-01 Theraptosis S.A. Peptides useful as dual caspase-2/-6 inhibitors and their biological applications

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Abstracts of the Annual Meeting of the Society for Neuroscience, Vol 43, 2013, V Vigneswara et al, Combined suppression of caspase-2 and caspase-6 protects retinal ganglion cells from apoptosis and promotes axon regeneration through CNTF-medicated JAK/STAT signalling pathway , 1-2, Program/Poster: *
Archives of Biochemistry and Biophysics, Vol 390, 2001, SW Rabkin, "Prevention of staurosporine-induced cell death in embryonic chick cardiomyocyte is more dependent on caspase-2 than caspase-3 inhibition and is independent of sphingomyelinase activation and ceramide generation", 119-127 *

Also Published As

Publication number Publication date
GB201404289D0 (en) 2014-04-23

Similar Documents

Publication Publication Date Title
Ma et al. Calpains mediate axonal cytoskeleton disintegration during Wallerian degeneration
Vigneswara et al. Combined suppression of CASP2 and CASP6 protects retinal ganglion cells from apoptosis and promotes axon regeneration through CNTF-mediated JAK/STAT signalling
EP2510098B1 (en) Methods and compositions for treating diseases, disorders or injury of the cns
US20110207795A1 (en) Smad7 inhibitor compositions and uses thereof
Wang et al. Intravitreal delivery of human NgR-Fc decoy protein regenerates axons after optic nerve crush and protects ganglion cells in glaucoma models
MX2008009355A (en) Therapeutic uses of inhibitors of rtp801.
JP5094395B2 (en) Treatment of gliosis, glial scar, inflammation, or axonal growth inhibition within the nervous system by modulating Eph receptors
CA2929669A1 (en) Compositions and methods for inhibiting nf-.kappa.b and sod-1 to treat amyotrophic lateral sclerosis
US10870852B2 (en) Compositions and methods for treating diabetic retinopathy
KR20190062363A (en) Therapeutic effects of Nurr1 and Foxa2 in inflammatory neurologic disorders by M1-to-M2 polarization of glial cells
Heiduschka et al. Aurintricarboxylic acid promotes survival and regeneration of axotomised retinal ganglion cells in vivo
US20140323549A1 (en) Methods and compositions for treating diseases, disorders or injury of the nervous system
Donnini et al. Prevention of ischemic brain injury by treatment with the membrane penetrating apoptosis inhibitor, TAT-BH4
GB2526767A (en) Combined inhibition of caspase-2 and caspase-6 in neuroprotection and axon regeneration
WO2015136255A1 (en) Combined inhibition of caspase-2 and caspase-6 in neuroprotection and axon regeneration
JPH09503512A (en) Methods for increasing neuronal survival and agents useful therefor
Shioya et al. Neutrophil elastase inhibitor suppresses IL-17 based inflammation of murine experimental colitis
US20220160838A1 (en) Compositions and methods for promoting islet viability and enhancing insulin secretion
US20160304879A1 (en) Double-Stranded RNA Compounds to CASP2 and Uses Thereof
TWI601821B (en) Treatment of neurodevelopmental disorders
CN108026530B (en) SiRNA and its use in methods and compositions for inhibiting expression of NRARP gene
JP2020527132A (en) Compositions and Methods for Treating Myelin Disorders
US20230167444A1 (en) Compositions and methods for intranasal treatment with double stranded rna
Patel et al. Postinfectious epilepsy
Dubowsky et al. Neurotrophic Therapy for ALS/MND

Legal Events

Date Code Title Description
WAP Application withdrawn, taken to be withdrawn or refused ** after publication under section 16(1)