WO2004037281A1 - Methods for neuroprotection - Google Patents

Methods for neuroprotection Download PDF

Info

Publication number
WO2004037281A1
WO2004037281A1 PCT/CA2003/001626 CA0301626W WO2004037281A1 WO 2004037281 A1 WO2004037281 A1 WO 2004037281A1 CA 0301626 W CA0301626 W CA 0301626W WO 2004037281 A1 WO2004037281 A1 WO 2004037281A1
Authority
WO
WIPO (PCT)
Prior art keywords
cntf
peptide
biologically
variant
active fragment
Prior art date
Application number
PCT/CA2003/001626
Other languages
French (fr)
Inventor
Mark A. Ozog
John Bechberger
Christian Naus
Original Assignee
The University Of British Columbia
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 The University Of British Columbia filed Critical The University Of British Columbia
Priority to AU2003275846A priority Critical patent/AU2003275846A1/en
Publication of WO2004037281A1 publication Critical patent/WO2004037281A1/en

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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the invention relates, in part, to methods for neuroprotection.
  • Acute and chronic neurodegeneration in the central nervous system is believed to result from a general process termed "excitotoxicity.”," which is mediated by the action of glutamate.
  • Glutamate is a negatively charged amino acid used for excitatory synaptic transmission in the mammalian nervous system. Although the concentration of glutamate can reach the millimolar range in nerve terminals (Waelsch, H., 1951), its extracellular concentration is maintained at a low level to prevent neurotoxicity. As early as 1957, Lucas and Newhouse
  • glutamate can be toxic to neurons if presented at a high concentration. Later, Olney and Sharpe (1969) implemented the term "excitotoxicity" to describe the cytotoxic effect that glutamate (and other such excitatory amino acids) has on neurons when applied at high dosages. Physiologically, high levels of glutamate can be achieved by excessive release, inhibition of uptake, or both. Normally, a low concentration of extracellular glutamate is maintained by both neurons and astrocytes. Neurons store glutamate in intracellular stores and regulate its release (Reagan, R.F., 1994).
  • Such therapies include glutamate release • . _ inhibitors, glutamate receptor antagonists, Ca 2+ channel blockers, GABA receptor agonists, gangliosides, neurotrophic factors, calpain inhibitors, caspase inhibitors, free radical scavengers, irnmuno- and cell metabolism modulators.
  • Gap junctions are proteineous channels that directly link the cytosol of adjacent cells and allow intercellular passage of molecules of M ⁇ ⁇ 1200 (Kumar and Gilula, 1996).
  • the structural unit of the gap junction is the connexon which itself consists of protein subunits termed connexins (Cxs)(Kumar and Gilula, 1996). At least eight different Cxs have been identified in the CNS. The connexins (Cxs) are designated according to relative molecular mass and numerous isoforms have been identified, including at least eight within the CNS (Dermietzel and Spray, 1993; Rozental et al., 2000; Willecke et al, 2002).
  • Cx43 the major Cxn expressed by astrocytes
  • astrocytes have also been reported to be in neurons (Dermietzel and Spray, 1993; Nadarajah et al., 1996; Alvarez-Maubecin et al, 2000; Nedergaard, 1994; Micevych and Abelson, 1991; Micevych et al, 1996).
  • Astrocytes play a supportive role for neurons by providing crucial metabolites, maintaining favourable extracellular levels of neurotransmitters and ions, and helping constitute the blood-brain barrier (Hatten and Mason, 1986).
  • astrocytes also have the ability to readily divide. Normal astrocytes are highly coupled by gap junctions and express primarily Cx43 and to a lesser extent Cxs 26, 30, 31.1 and 40 (Dermietzel et al, 2000).
  • gap junctions can be altered by short-term and long-term regulation (reviewed by Giaume and McCarthy, 1996; Rouach and Giaume, 2001).
  • Short-term regulation occurs within milliseconds to minutes and is the result of a change in the biophysical properties of the channel.
  • Such channel gating is usually caused by post-translational processing such as phosphorylation or channel blocking.
  • Long-term regulation occurs over hours or days and consists of the formation or disappearance of gap junction channels as a result of changes to gene transcription and post-translational processes. While numerous agents have been identified to modulate short-term gap junction regulation (reviewed by Dhein, 1998), long-term regulators have not been extensively studied.
  • Gap junctional coupling can be impaired/blocked by administering various agents including carbenoxolone (3 -hydroxy- l l-oxoolean-12-en-30-oicacid 3 hemisuccinate; CBX) and 18 ⁇ -glycyrrhetinic acid (3-hydroxy-ll-oxo-18,20-olean-12-en-29-oic acid; AGA).
  • CBX is a water-soluble synthetic gap junction blocker and has an inactive analog, glycyrrhizic acid (GZA), which is a derivative of Glycyrrhiza glabra.
  • GZA glycyrrhizic acid
  • AGA is also derived from C glabra and also blocks gap junctions; the exact mechanism to which AGA and CBX block the junctions has not yet been deciphered. Previous studies have reported that these blocking agents have no effect on cell viability if presented at concentrations less than 100 ⁇ M.
  • Cx43 is upregulated in astrocytes treated with transforming growth factor- ⁇ l (Robe et al, 2000) but downregulated by both interleukin (IL)-l ⁇ (John et al, 1999) and transforming growth factor- ⁇ 3 (Reuss et al., 1998).
  • Fibroblast growth factor-2 has opposing effects on cells of the CNS, increasing Cx43 expression in neurons (SiuYi et al, 2001) but decreasing this gap junction component in astrocytes (Reuss et al., 1998; Reuss et al, 2000).
  • Prior research suggests that Cx43 is up-regulated following a CNS insult (Enkvist and McCarthy, 1994; Hossain et al, 1994).
  • Gap junctions could have a direct protective effect, allowing the astrocytic syncytium to buffer extracellular space from cytotoxic levels of metabolites and ions (such as glutamate and K + ).
  • gap junctions could promote the viability of energy- exhausted cells by allowing essential molecules (e.g. ATP and glucose) to move into areas of high demand.
  • essential molecules e.g. ATP and glucose
  • gap junctions could permit the movement of potentially toxic metabolites into cells that are already compromised by the insult.
  • Ciliary neurotrophic factor is a cytokine normally found within the cytosol of astrocytes. CNTF was originally identified, purified and cloned based upon its ability to support the in viiro survival of parasympathetic neurons (Barbin et al, 1984; Lin et al, 1989; Masiakowski et al., 1991; Stockli et al, 1989).
  • CNTF is a member of the IL-6 family that includes IL-6, IL-11, oncostatin M, cardiotrophin-1 and leukemia inhibitory factor (LIF)(Ip and Yancopoulos, 1992; Lin et al, 1989; Patterson, 1992; Stockli et al, 1989); all of these family members share the signaling receptor system of glycoprotein 130 (gpl30) and LIF receptor- ⁇ (LIFR ⁇ ) to initiate intracellular signaling cascades.
  • LIF leukemia inhibitory factor
  • the expression of CNTF is almost exclusively restricted to nervous tissue, where it is normally produced by astrocytes in the CNS and Schwann cells in the peripheral nervous system (Stockli et al, 1991).
  • CNTF is considered a "brain injury" cytokine based on the findings that: 1) astrocytes express and retain CNTF as a cytosolic protein which is only released following brain disturbances (Stockli et al, 1989; Stockli et al, 1991; Rudge et al., 1994a; Rudge et al.,
  • the CNTF specific receptor, CNTFR-alpha or CNTFR ⁇ is located on neurons (Ip et al, 1993a; Kahn et al, 1997; Maclennan et al, 1996) and post-insulted (reactive) astrocytes (Ip et al., 1993b; Rudge et al, 1995); and 3) expression of both CNTF and CNTFR ⁇ is upregulated following disturbances to the CNS (Asada et al., 1995; Asada et al, 1996; Duberley et al, 1995; Ip et al, 1993b; Lee et al, 1997; Lin et al, 1998; Nieto-Sampedro et al, 1982; Park et al, 2000; Rudge et al, 1994a; Wen et al, 1995a).
  • CNTFR ⁇ Upon release by injured astrocytes, CNTF binds to CNTFR ⁇ and subsequently recruits gpl30 and LIFR ⁇ through which a cascade of several signal transduction pathways is initiated.
  • CNTFR ⁇ can be cleaved (i.e., by PLC) from expressing cells and, as a soluble form, can bind CNTF and activate cellular pathways.
  • CNTF may bind its receptor (CNTFR ⁇ ; soluble or membrane bound form) and subsequently activate a cascade of signal transduction pathways.
  • CNTFR ⁇ soluble or membrane bound form
  • the predominant pathways activated by CNTF are the mitogen-activated protein kinase/extracellular signal- regulated kinase (MAPK/ERK) pathway and the j anus kinases/signal transducers of activated transcription (JAK/STAT) pathway (reviewed by Monville et al, 2001). Both MAPK/ERK and JAK/STAT pathways lead to direct transcriptional activation of various genes (Symes et al, 1994).
  • CNTFR ⁇ lacks ⁇ ransmembrane and cytoplasmic domains and is instead anchored to the cell surface by a glycosyl-phosphotidylinositol linkage (Davis, et al. 1991). This linkage can be cleaved by phospholipases and release the receptor as a soluble protein. Although the soluble receptor is functional, it requires CNTF to be released into the extracellular space by dying or compromised cells to which it can bind and subsequently activate the ubiquitously expressed ⁇ components of the receptor. Davis, et al. (1993b) demonstrated that administration of both the soluble receptor and CNTF to various hematopoietic cell lines mimicked the effect of leukemia inhibitory factor (LIF) on these cells.
  • LIF leukemia inhibitory factor
  • the hematopoietic cell lines were not normally responsive to CNTF alone and did not express the receptor. However, only hematopoietic cell lines expressing gpl30 and the ⁇ receptor for LIF responded to the combination of CNTF and its soluble receptor. The authors speculated that CNTFR ⁇ may participate in interactions between the nervous system and hematopoietic and other systems. Similarly, combination of the receptor and CNTF has been shown to promote macrophage chemotaxis in a manner similar to interleukin 6 (Kobayashi, H. & Mizisin, A.P., 2000).
  • This invention arises from the discovery that co-administration of a CNTF peptide and at a peptide that acts as a receptor for CNTF provides a neuroprotective effect on CNS tissue exceeding the neuroprotective effect of CNTF alone. These effects appear to be brought about by the action of a combination of the receptor and CNTF on mature or un-reactive astrocytes, or alternatively, neoplastic cells of astrocytic origin.
  • Neuroprotection provided by this invention includes protection from damage resulting from neural injury or insult and from neurotoxicity, including excitotoxicity.
  • neuroprotection provided by this invention will be useful in the treatment of acute and chronic neurodegenerative disorders that may involve excitotoxicity, for example glutamate excitotoxicity, including stroke/ischemia, trauma, epilepsy, Huntington's Disease, amyotrophic lateral sclerosis and hypoglycemic encephalopathy.
  • Neuroprotection provided by this invention may be brought about upon injured or diseased tissue or in a preventative fashion during or prior to events expected to lead to a neural insult.
  • the invention provides methods for providing neuroprotection; for inhibiting cell degeneration or cell death; for treatment or prophylaxis of a neurodegenerative disease; or for ameliorating the cytotoxic effect of a compound (for example, a excitatory amino acid such as glutamate; a toxin; or a prophylactic or therapeutic compound that exerts a cytotoxic side effect) in a subject in need thereof, by administering to the subject an effective amount of a CNTF peptide or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding the CNTF peptide or a biologically-active fragment or variant thereof; and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof.
  • the methods of the invention include protection against excitotoxicity, for example glutamate excito
  • the subject for example, a human, may be suffering from neural insult or injury; or may be suffering from a condition selected from substance abuse, trauma, stroke, ischemia, Huntington's disease, Alzheimer's disease, Parkinson's disease, prion disease, variant Creutzfeld- Jakob disease, amyotrophic or hypoglycemic encephalopathy; or may be undergoing surgery or other intervention.
  • the subject may have a pre-existing condition which would benefit by neuroprotection or the patient may be treated to reduce deleterious effects of a concomitant or subsequent neural injury, such as may occur during surgery or other intervention.
  • the peptide that acts as a receptor for CNTF is soluble (e.g., soluble CNTFR ⁇ ).
  • the peptides are administered as a complex.
  • the invention also provides the use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof for neuroprotection; for modulation of cell degeneration or cell death; for treatment or prophylaxis of a neurodegenerative disorder; or for ameliorating the cytotoxic effect of a compound.
  • the invention also provides the use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof for preparation of a medicament for neuroprotection; modulation of cell degeneration or cell death; treatment or prophylaxis of a neurodegenerative disorder; or ameliorating the cytotoxic effect of a compound.
  • compositions including a CNTF peptide or a biologically- active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof for neuroprotection; modulation of cell degeneration or cell death; treatment or prophylaxis of a neurodegenerative disorder; or ameliorating the cytotoxic effect of a compound.
  • the uses of the invention include protection against excitotoxicity, for example glutamate excitotoxicity.
  • the invention provides a method of screening for a neuroprotective compound, by providing a first system including CNTF or a biologically-active fragment or variant thereof; providing a second system including a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof; providing a third system including CNTF or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof; contacting the first and second systems with a test compound; and determining whether the test compound modulates neuroprotection in the first or second system when compared to the third system.
  • the invention also provides a method of screening for a neuroprotective compound, by providing a system including CNTF or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof; contacting the system with a test compound; and determining whether the test compound modulates neuroprotection in the presence or absence of a gap junction blocker.
  • the invention also provides a method of screening for a nucleic acid molecule that modulates neuroprotection, by providing a neuronal or neuronal-associated cell; contacting the neuronal or neuronal-associated cell with CNTF or a biologically-active fragment or variant thereof; a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof; or a combination comprising the CNTF and the peptide that acts as a receptor for CNTF; and determining the level of nucleic acid molecule expression (for example, by a microarray), where a change in the level of nucleic acid molecule expression in the combination, when compared to the level of nucleic acid molecule expression in either CNTF or the CNTF receptor systems alone, is indicative of a nucleic acid molecule that modulates neuroprotection.
  • the neuroprotection may include protection of neuronal cells or neuronal-associated cells from injury or degeneration or increasing the expression level of a connexin (e.g. Cx43).
  • the system(s) may include neuronal or neuronal associated cells.
  • this invention also provides a method for treatment of a CNS tumor in a patient in need thereof, comprising administering to the patient an effective amount of the aforementioned complex or a pharmaceutical composition comprising the aforementioned complex.
  • This separate aspect also provides the use of a complex of a peptide having the activity of CNTF and a peptide having the function of a receptor for CNTF the treatment of neoplastic disorders.
  • This separate aspect also provides the use of the aforementioned complex for preparation of medicaments for treatment of neoplastic disorders.
  • This anti-neoplastic aspect of the invention is distinct from the neuroprotection aspect.
  • the separate aspect of this invention also provides a method for determining the susceptibility of a neoplastic cell to suppression by the aforementioned complex, comprising determining whether the cell expresses CNTF.
  • This method may further comprise determining whether the cell expresses CNTFR ⁇ .
  • the method may also further comprise isolating and culturing neoplastic cells to be tested, and may further comprise obtaining a sample of tissue from a subject comprising such cells to be tested.
  • the determining may be by any means for detecting the presence of, or quantification of expression, including detection and/or measurement of mRNA or detection and/or quantification of expressed protein.
  • Such detection and/or quantification may involve nucleic acid probing and detection of nucleic acids hybridized to a suitable probe or amplification of target nucleic acids using suitable primers.
  • Detection and/or quantification of expressed protein may involve immunological procedures whereby an antibody to the protein is employed in procedures known in the art.
  • Medicaments and pharmaceutical compositions for use in this invention will comprise one or more peptides having the activity of CNTF and one or more peptides capable of functioning as a CNTF receptor.
  • the peptide functioning as the receptor is a soluble peptide capable of binding CNTF.
  • Pharmaceutical compositions and medicaments of this invention may also comprise one or more pharmaceutically acceptable diluents, carriers, excipients or the like including those known in the art for formulation and facilitation of delivery of peptides to target cells or tissues, including CNS tissue.
  • neurodegenerative disorder is meant a disorder that is characterized by the death or loss of function of neuronal cells, also known as neurons.
  • a neurodegenerative disorder is characterized by the death or loss of function of neuronal- associated cells, such as astrocytes or other glial cells. The cell death may occur by apoptosis or by necrosis.
  • a neurogenerative disorder includes, without limitation, substance abuse, trauma, stroke, ischemia, Huntington's disease, Alzheimer's disease, Parkinson's disease, prion diseases, such as variant Creutzfeld- Jakob, amyotrophic lateral sclerosis (ALS), olivopontocerebellar atrophy, epilepsy, seizures, and hypoglycemic encephalopathy.
  • a neurodegenerative disorder also includes any disorder or condition that would benefit from neuroprotection.
  • Neuronal cells such as neurons
  • neuroprotection refers to protection of any nerve tissue, including neurons and neuronal-associated cells (such as astrocytes or other glial cells) from injury or degeneration.
  • the injury or degeneration may lead to cell death or loss of function.
  • the injury or degeneration may result from a general process termed "excitotoxicity,” which is mediated by the action of glutamate.
  • the injury or degeneration may be due to physical injury or trauma; a neurodegenerative disorder, including a hereditary genetic disorder; action of a compound, for example, a toxin or a drug, such as a therapeutic or prophylactic; or any other factor that results in nerve tissue injury or degeneration.
  • the neuroprotection may extend to at least a portion of affected or injured nerve tissue. For example, in stroke, while neurons within the core area of the ischemic insult may be unsalvageable, neurons within the surrounding penumbra have the potential to be rescued, since most neurons within the penumbra do not succumb to the ischemic insult until days following the stroke, and may benefit from the neuroprotective compositions of the invention.
  • the compounds including test compounds, or combinations of the invention, for example, a CNTF/soluble CNTF receptor combination, exhibit any integer between 10% and 90%, for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, or over 100%, neuroprotection when compared to no treatment, or treatment with either CNTF or soluble CNTF receptor alone.
  • “Modulating” or “modulates” means changing, by either increase or decrease. The increase or decrease may be a change of any integer value between 10% and 90%, or of any integer value between 30% and 60%, or may be over 100%, when compared with a control or reference sample or compound.
  • test compound is any naturally-occurring or artificially-derived chemical compound.
  • Test compounds may include, without limitation, peptides, polypeptides, synthesised organic molecules, naturally occurring organic molecules, and nucleic acid molecules.
  • a test compound can "compete" with a known compound or combination such as a CNTF/soluble CNTF receptor combination by, for example, interfering with upregulation of connexin protein or mRNA expression levels, e.g., Cx43, Cxs 26, Cxs 30, Cxs 31.1, or Cxs 40 levels, by interfering with the action of gap junction blockers, or by interfering with any biological response induced by the known combination, such as neuroprotection.
  • a test compound can exhibit any value between 10% and 200%), or over 500%, modulation when compared to a CNTF/soluble CNTF receptor combination, or other reference compound.
  • a test compound may exhibit at least any positive or negative integer from 10% to 200% modulation, or at least any positive or negative integer from 30% to 150% modulation, or at least any positive or negative integer from 60% to 100% modulation, or any positive or negative integer over 100% modulation.
  • a compound that is a negative modulator will in general decrease modulation relative to a known compound, while a compound that is a positive modulator will in general increase modulation relative to a known compound.
  • contacting is meant to submit an animal, cell, lysate, extract, molecule derived from a cell, or synthetic molecule to a test compound.
  • determining is meant analysing the effect of a test compound on the test system.
  • the means for analysing may include, without limitation, polymerase chain reaction, intercellular coupling assays, immunological assays (e.g, immunoprecipitation, immunofluorescence, ELISA, Western blotting), ultrastructural analysis, histological analysis, kinase assays, cell death assays, Northern or Southern blotting, gene array assays, animal models, or any other methods described herein or known to those skilled in the art.
  • immunological assays e.g, immunoprecipitation, immunofluorescence, ELISA, Western blotting
  • ultrastructural analysis e.g, histological analysis, kinase assays, cell death assays, Northern or Southern blotting, gene array assays, animal models, or any other methods described herein or known to those skilled in the art.
  • nucleic acid or “nucleic acid molecule” encompass both RNA (plus and minus strands) and DNA, including cDNA, genomic DNA, and synthetic (e.g., chemically synthesized) DNA.
  • the nucleic acid may be double-stranded or single-stranded. Where single-stranded, the nucleic acid may be the sense strand or the antisense strand.
  • a nucleic acid molecule may be any chain of two or more covalently bonded nucleotides, including naturally occurring or non-naturally occurring nucleotides, or nucleotide analogs or derivatives.
  • RNA is meant a sequence of two or more covalently bonded, naturally occurring or modified ribonucleotides.
  • RNA is phosphorothioate RNA.
  • DNA is meant a sequence of two or more covalently bonded, naturally occurring or modified deoxyribonucleotides.
  • cDNA is meant complementary or copy DNA produced from an RNA template by the action of RNA-dependent DNA polymerase (reverse transcriptase).
  • a cDNA clone means a duplex DNA sequence complementary to an RNA molecule of interest, carried in a cloning vector.
  • nucleic acids e.g., DNA or RNA
  • nucleic acids e.g., DNA or RNA
  • each nucleotide in a nucleic acid molecule need not form a matched Watson-Crick base pair with a nucleotide in an opposing complementary strand to form a duplex.
  • a nucleic acid molecule is "complementary" to another nucleic acid molecule if it hybridizes, under conditions of high stringency, with the second nucleic acid molecule.
  • the level of nucleic acid molecule expression may be measured by any technique known in the art, for example, by quantification of hybridization signals.
  • a change in the level of nucleic acid molecule expression may a change of any value between 10% and 200%, for example a change of 10%, 30%, 50%, 100%, or 150%, or over 500%, when compared to a reference system or compound.
  • a "microarray” generally refers to a high density nucleic acid molecule array, which may for example be used to monitor the presence or level of expression of a large number of genes or for detecting sequence variations, mutations and polymorphisms. Microarrays generally require a solid support (for example, nylon, glass, ceramic, plastic, etc.) to which the nucleic acid molecules are attached in a specified 2-dimensional arrangement, such that the pattern of hybridization to a probe is easily determinable.
  • a solid support for example, nylon, glass, ceramic, plastic, etc.
  • the invention provides, in part, methods for neuroprotection using a "Complex" or a combination of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that as a receptor for CNTF, e.g., CNTFR-alpha, or a biologically-active fragment or variant thereof.
  • the Complex can increase cellular communication among CNS cells, and therefore increases the resilience of CNS cells to insults or injury.
  • Neuroprotection using the Complex or combination of the invention is more effective than that achieved in the absence of treatment (i.e., without applying exogenous agents) or by treatment with CNTF alone, or with soluble CNTF receptor alone.
  • Compounds for use in this invention include, without limitation, a peptide having the activity of CNTF; a peptide having the activity or function of a CNTF receptor, e.g., CNTFR- alpha; and combinations thereof.
  • Narious peptides and sequences of peptides having the activity of C ⁇ TF are known in the art and additional variants may be prepared by persons of skill in the art or as described herein. Examples of prior literature describing C ⁇ TF and homologous peptides include WO 91/04316 and United States Patents 4,997,929, 5,011,914 and 5,426,177.
  • Narious peptides and sequences of peptides having the activity of a C ⁇ TF receptor, including its soluble portion, are known in the art and additional variants may be prepared by persons of skill in the art or as described herein.
  • Examples of prior literature describing C ⁇ TF receptors, including their soluble portions and homologous peptides include Davis, et al. (1991); and United States Patents 5,426,177 and 5,849,897.
  • a soluble peptide having the activity of a CNTF receptor may be obtained commercially, for example, from R&D Systems, Minneapolis, MN, USA.
  • a soluble peptide having the activity of a CNTF receptor may also be obtained by, for example, cleaving the membrane linkage of a CNTF receptor using a phospholipase, or by recombinant techniques whereby a peptide having the activity of a CNTF receptor is mutated so that it is no longer capable of associating with membranes.
  • a peptide having the activity of a CNTF receptor is capable of binding CNTF.
  • a peptide having the activity of a CNTF receptor is also capable of binding gpl30 and/or the LIF receptor.
  • Antibodies to the receptor are known, including those described in United States Patent 5,892,003.
  • the CNTF and/or CNTF receptor may be human, or may be derived from any other animal, such as mouse, rat, rabbit, chicken, pig, or monkey.
  • the invention excludes polypeptide compounds composed of a fusion between CNTF and a CNTF receptor, for example, a full length CNTF receptor.
  • the invention includes polypeptides including CNTF fused with a soluble CNTF receptor.
  • a "protein,” “peptide” or “polypeptide” is any chain of two or more amino acids, including naturally occurring or non-naturally occurring amino acids or amino acid analogues, regardless of post-translational modification (e.g., glycosylation or phosphorylation).
  • amino acid sequence may include peptides or proteins that have abnormal linkages, cross links and end caps, non-peptidyl bonds or alternative modifying groups. Such modified peptides are also within the scope of the invention.
  • modifying group is intended to include structures that are directly attached to the peptidic structure (e.g., by covalent coupling), as well as those that are indirectly attached to the peptidic structure (e.g., by a stable non-covalent association or by covalent coupling to additional amino acid residues, or mimetics, analogues or derivatives thereof, which may flank the core peptidic structure).
  • the modifying group can be coupled to the amino-terminus or carboxy-terminus of a peptidic structure, or to a peptidic or peptidomimetic region flanking the core domain.
  • the modifying group can be coupled to a side chain of at least one amino acid residue of a peptidic structure, or to a peptidic or peptido- mimetic region flanking the core domain (e.g., through the epsilon amino group of a lysyl residue(s), through the carboxyl group of an aspartic acid residue(s) or a glutamic acid residue(s), through a hydroxy group of a tyrosyl residue(s), a serine residue(s) or a threonine residue(s) or other suitable reactive group on an amino acid side chain).
  • Modifying groups covalently coupled to the peptidic structure can be attached by means and using methods well known in the art for linking chemical structures, including, for example, amide, alkylamino, carbamate or urea bonds.
  • a "biologically-active fragment" of CNTF or a peptide having the activity or function of a CNTF receptor, e.g., CNTFR-alpha includes an amino acid sequence found in a naturally-occurring CNTF or a soluble CNTF receptor peptide that is capable of neuroprotection as described herein or known to those of ordinary skill in the art.
  • a "variant" of CNTF or a peptide having the activity or function of a CNTF receptor includes a modification, for example, by deletion, addition, or substitution, of an amino acid sequence found in a naturally-occurring CNTF or a soluble CNTF receptor peptide that is capable of neuroprotection as described herein or known to those of ordinary skill in the art.
  • Compounds can be prepared by, for example, replacing, deleting, or inserting an amino acid residue at any position of CNTF or a peptide having the activity or function of a CNTF receptor, e.g., CNTFR-alpha, with other conservative amino acid residues, i.e., residues having similar physical, biological, or chemical properties, and screening for the ability of the compound to modulate neuroprotection.
  • a CNTF receptor e.g., CNTFR-alpha
  • polypeptides of the present invention also extend to biologically equivalent peptides that differ from a portion of the sequence of the polypeptides of the present invention by conservative amino acid substitutions.
  • substitutions refers to the substitution of one amino acid for another at a given location in the peptide, where the substitution can be made without substantial loss of the relevant function.
  • substitutions of like amino acid residues can be made on the basis of relative similarity of side-chain substituents, for example, their size, charge, hydrophobicity, hydrophilicity, and the like, and such substitutions may be assayed for their effect on the function of the peptide by routine testing.
  • amino acids means those L-amino acids commonly found in naturally occurring proteins, D-amino acids and such amino acids when they have been modified. Accordingly, amino acids of the invention may include, for example: 2-Aminoadipic acid; 3-Aminoadipic acid; beta-Alanine; beta-Aminopropionic acid; 2-Aminobutyric acid; 4- Aminobutyric acid; piperidinic acid; 6-Aminocaproic acid; 2-Aminoheptanoic acid; 2- Aminoisobutyric acid; 3-Aminoisobutyric acid; 2-Aminopimelic acid; 2,4 Diaminobutyric acid; Desmosine; 2,2'-Diaminopimelic acid; 2,3-Diaminopropionic acid; N-Ethylglycine; N- Ethylasparagine; Hydroxylysine; allo-Hydroxylysine; 3-Hydroxyproline; 4-Hydr
  • conserved amino acid substitutions may be made where an amino acid residue is substituted for another having a similar hydrophilicity value (e.g., within a value of plus or minus 2.0, or plus or minus 1.5, or plus or minus 1.0, or plus or minus 0.5), where the following may be an amino acid having a hydropathic index of about -1.6 such as Tyr (-1.3) or Pro (-1.6) are assigned to amino acid residues (as detailed in United States Patent No.
  • conservative amino acid substitutions may be made where an amino acid residue is substituted for another having a similar hydropathic index (e.g., within a value of plus or minus 2.0, or plus or minus 1.5, or plus or minus 1.0, or plus or minus 0.5).
  • each amino acid residue may be assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics, as follows: He (+4.5); Nal (+4.2); Leu (+3.8); Phe (+2.8); Cys (+2.5); Met (+1.9); Ala (+1.8); Gly (-0.4); Thr (-0.7); Ser (-0.8); Trp (-0.9); Tyr (-1.3); Pro (-1.6); His (-3.2); Glu (-3.5); Gin (-3.5); Asp (-3.5); Asn (-3.5); Lys (-3.9); and Arg (-4.5).
  • conservative amino acid substitutions may be made using publicly available families of similarity matrices (Altschul, S.F. 1991. "Amino acid substitution matrices from an information theoretic perspective.” Journal of Molecular Biology, 219: 555- 665; Dayhoff, M.O., Schwartz, R.M., Orcutt, B.C. 1978. "A model of evolutionary change in proteins.” In “Atlas of Protein Sequence and Structure” 5(3) M.O.
  • conservative amino acid substitutions may be made where an amino acid residue is substituted for another in the same class, where the amino acids are divided into non-polar, acidic, basic and neutral classes, as follows: non-polar: Ala, Nal, Leu, He, Phe, Trp, Pro, Met; acidic: Asp, Glu; basic: Lys, Arg, His; neutral: Gly, Ser, Thr, Cys, Asn, Gin, Tyr.
  • Conservative amino acid changes can include the substitution of an L-amino acid by the corresponding D-amino acid, by a conservative D-amino acid, or by a naturally-occurring, non- genetically encoded form of amino acid, as well as a conservative substitution of an L-amino acid.
  • Naturally-occurring non-genetically encoded amino acids include beta-alanine, 3-amino- propionic acid, 2,3-diamino propionic acid, alpha-aminoisobutyric acid, 4-amino-butyric acid, N-methylglycine (sarcosine), hydroxyproline, ornithine, citrulline, t-butylalanine, t- butylglycine, N-methylisoleucine, phenylglycine, cyclohexylalanine, norleucine, norvaline, 2- napthylalanine, pyridylalanine, 3-benzothienyl alanine, 4-chlorophenylalanine, 2- fluorophenylalanine, 3-fluorophenylalanine, 4-fluorophenylalanine, penicillamine, 1,2,3,4- tetrahydro-isoquinoline-3-carboxylix acid, beta-2-thienylalan
  • conservative amino acid changes include changes based on considerations of hydrophilicity or hydrophobicity, size or volume, or charge.
  • Amino acids can be generally characterized as hydrophobic or hydrophilic, depending primarily on the properties of the amino acid side chain.
  • a hydrophobic amino acid exhibits a hydrophobicity of greater than zero, and a hydrophilic amino acid exhibits a hydrophilicity of less than zero, based on the normalized consensus hydrophobicity scale of Eisenberg et al(J. Mol. Bio. 179:125-142, 184).
  • hydrophobic amino acids include Gly, Ala, Phe, Nal, Leu, He, Pro, Met and Trp, and genetically encoded hydrophilic amino acids include Thr, His, Glu, Gin, Asp, Arg, Ser, and Lys.
  • ⁇ on-genetically encoded hydrophobic amino acids include t-butylalanine, while non-genetically encoded hydrophilic amino acids include citrulline and homocysteine.
  • Hydrophobic or hydrophilic amino acids can be further subdivided based on the characteristics of their side chains.
  • an aromatic amino acid is a hydrophobic amino acid with a side chain containing at least one aromatic or heteroaromatic ring, which may contain one or more substituents such as -OH, -SH, -C ⁇ , -F, -CI, -Br, -I, - ⁇ O 2 , -NO, - NH 2 , -NHR, -NRR, -C(O)R, -C(O)OH, -C(O)OR, -C(O)NH 2 , -C(O)NHR, -C(O)NRR, etc., where R is independently (C ⁇ -C 6 ) alkyl, substituted (C ⁇ -C 6 ) alkyl, (C ⁇ -C 6 ) alkenyl, substituted (C ⁇ -C 6 ) alkenyl, (C ⁇ -C 6 ) alkynyl
  • Aromatic amino acids include Phe, Tyr, and Trp, while non-genetically encoded aromatic amino acids include phenylglycine, 2-napthylalanine, beta-2- thienylalanine, 1, 2,3 ,4-tetrahydro-isoquinoline-3 -carboxylic acid, 4-chlorophenylalanine, 2- fluorophenylalanine3 -fluorophenylalanine, and 4-fluorophenylalanine.
  • An apolar amino acid is a hydrophobic amino acid with a side chain that is uncharged at physiological pH and which has bonds in which a pair of electrons shared in common by two atoms is generally held equally by each of the two atoms (i.e., the side chain is not polar).
  • Genetically encoded apolar amino acids include Gly, Leu, Nal, He, Ala, and Met, while non- genetically encoded apolar amino acids include cyclohexylalanine.
  • Apolar amino acids can be further subdivided to include aliphatic amino acids, which is a hydrophobic amino acid having an aliphatic hydrocarbon side chain.
  • Genetically encoded aliphatic amino acids include Ala, Leu, Nal, and He, while non-genetically encoded aliphatic amino acids include norleucine.
  • a polar amino acid is a hydrophilic amino acid with a side chain that is uncharged at physiological pH, but which has one bond in which the pair of electrons shared in common by two atoms is held more closely by one of the atoms.
  • Genetically encoded polar amino acids include Ser, Thr, Asn, and Gin, while non-genetically encoded polar amino acids include citrulline, ⁇ -acetyl lysine, and methionine sulfoxide.
  • An acidic amino acid is a hydrophilic amino acid with a side chain pKa value of less than 7. Acidic amino acids typically have negatively charged side chains at physiological pH due to loss of a hydrogen ion. Genetically encoded acidic amino acids include Asp and Glu. A basic amino acid is a hydrophilic amino acid with a side chain pKa value of greater than 7. Basic amino acids typically have positively charged side chains at physiological pH due to association with hydronium ion. Genetically encoded basic amino acids include Arg, Lys, and His, while non-genetically encoded basic amino acids include the non-cyclic amino acids ornithine, 2,3,-diaminopropionic acid, 2,4-diaminobutyric acid, and homoarginine.
  • amino acids can be classified based on known behaviour and or characteristic chemical, physical, or biological properties based on specified assays or as compared with previously identified amino acids.
  • Amino acids can also include bifunctional moieties having amino acidlike side chains.
  • Conservative changes can also include the substitution of a chemically derivatised moiety for a non-derivatised residue, by for example, reaction of a functional side group of an amino acid.
  • substitutions can include compounds whose free amino groups have been derivatised to amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t- butyloxycarbonyl groups, chloroacetyl groups or formyl groups.
  • free carboxyl groups can be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides, and side chains can be derivatized to form O-acyl or O-alkyl derivatives for free hydroxyl groups or N-im-benzylhistidine for the imidazole nitrogen of histidine.
  • Peptide analogues also include amino acids that have been chemically altered, for example, by methylation, by amidation of the C-terminal amino acid by an alkylamine such as ethylamine, ethanolamine, or ethylene diamine, or acylation or methylation of an amino acid side chain (such as acylation of the epsilon amino group of lysine).
  • a substituted amide for example, groups of the formula -C(O)-NR, where R is (C ⁇ -C 6 ) alkyl, (C ⁇ -
  • the compound can be covalently linked, for example, by polymerisation or conjugation, to form homopolymers or heteropolymers.
  • Spacers and linkers typically composed of small neutral molecules, such as amino acids that are uncharged under physiological conditions, can be used. Linkages can be achieved in a number of ways. For example, cysteine residues can be added at the peptide termini, and multiple peptides can be covalently bonded by controlled oxidation. Alternatively, heterobifunctional agents, such as disulfide/amide forming agents or thioether/amide forming agents can be used.
  • the compound can also be linked to a another compound that can modulate a neuroprotective response.
  • the compound can also be constrained, for example, by having cyclic portions.
  • Peptides or peptide analogues can be synthesised by standard chemical techniques, for example, by automated synthesis using solution or solid phase synthesis methodology.
  • test extracts or compounds are not critical to the method(s) of the invention. Accordingly, virtually any number of chemical extracts or compounds can be screened using the exemplary methods described herein. Examples of such extracts or compounds include, but are not limited to, plant-, fungal-, prokaryotic- or animal-based extracts, fermentation broths, and synthetic compounds, as well as modification of existing compounds. Numerous methods are also available for generating random or directed synthesis (e.g., semi-synthesis or total synthesis) of any number of chemical compounds, including, but not limited to, saccharide-, lipid-, peptide-, and nucleic acid-based compounds. Synthetic compound libraries are commercially available.
  • libraries of natural compounds in the form of bacterial, fungal, plant, and animal extracts are commercially available from a number of sources, including Biotics (Sussex, UK), Xenova (Slough, UK), Harbor Branch Oceanographic Institute (Ft. Pierce, FL, USA), and PharmaMar, MA, USA.
  • natural and synthetically produced libraries of, for example, candidate polypeptides are produced, if desired, according to methods known in the art, e.g., by standard extraction and fractionation methods.
  • any library or compound is readily modified using standard chemical, physical, or biochemical methods.
  • Compounds identified as being of therapeutic, prophylactic, diagnostic, or other value may be subsequently analyzed using a murine stroke model, or any other animal model suitable for neuroprotection studies, such as animal models of neurodegenerative disease (see, for example, Jankowsky JL et al, 2002, Transgenic mouse models of neurodegenerative disease: opportunities for therapeutic development, Curr Neurol Neurosci Rep. Sep;2(5):457-64; Hodgson et al. (1999) Neuron 23, 181-192).
  • compositions for example, nucleic acid molecules, small molecules, peptides, or peptide analogues
  • a liposome for example, an adjuvant, or any pharmaceutically acceptable carrier
  • treatment with a compound according to the invention may be combined with more traditional and existing therapies for providing neuroprotection, or for treating or prevention a neurodegenerative disorder.
  • the CNTF peptide, or biologically-active fragment or variant thereof may be administered in the same formulation as the soluble peptide having the activity or function of a CNTF receptor, e.g., CNTFR-alpha, or biologically-active fragment or variant thereof, or may be administered as separate formulations. If administered in the same formulation, in some embodiments, the CNTF peptide, or biologically-active fragment or variant thereof, and the soluble peptide having the activity or function of a CNTF receptor, e.g., CNTFR-alpha, or biologically-active fragment or variant thereof, may be administered as a complex, for example, as a covalently bound complex, or a non-covalently bound complex.
  • a "complex" refers to a combination in which the relevant ingredients are capable interacting, such as binding of CNTF to a soluble peptide or compound having the activity or function of a CNTF receptor.
  • compositions may be employed to provide suitable formulations or compositions to administer the compounds to subjects suffering from or presymptomatic for a neurodegenerative disorder, or in need of neuroprotection.
  • Any appropriate route of administration may be employed, for example, parenteral, intravenous, subcutaneous, intramuscular, intracranial, intraorbital, ophthalmic, intraventricular, intracapsular, intraspinal, intrathecal, intracisternal, intraperitoneal, intranasal, aerosol, or oral administration.
  • Therapeutic formulations maybe in the form of liquid solutions or suspensions; for oral administration, formulations may be in the form of tablets or capsules; and for intranasal formulations, in the form of powders, nasal drops, or aerosols.
  • the compounds of the invention are delivered directly or indirectly to CNS tissue.
  • Methods for formulation and delivery of peptides to CNS tissue are known, including methods for administering CNTF to the brain as described in United States Patent application 10/073,658 published June 13, 2002.
  • Formulations for parenteral administration may, for example, contain excipients, sterile water, or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated napthalenes.
  • Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds.
  • parenteral delivery systems for modulatory compounds include ethylene- vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
  • Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel.
  • the compounds are administered to an individual in an amount sufficient to stop or slow neurodegeneration or to provide neuroprotection, depending on the disorder.
  • An "effective amount" of a compound according to the invention includes a therapeutically effective amount or a prophylactically effective amount.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as neuroprotection, or the inhibition or prevention of neurodegeneration.
  • a therapeutically effective amount of a compound may vary- according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects.
  • a prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result, such as neuroprotection, or the inhibition or prevention of neurodegeneration.
  • a prophylactic dose is used in subjects prior to or at an earlier stage of disease, so that a prophylactically effective amount may be less than a therapeutically effective amount.
  • a preferred range for therapeutically or prophylactically effective amounts of a compound may be any integer from 0.1 nM-O.lM, 0.1 nM-0.05M, 0.05 nM-15 ⁇ M or 0.01 nM-10 ⁇ M. It is to be noted that dosage values may vary with the severity of the condition to be alleviated.
  • dosage regimens maybe adjusted over time according to the individual need and the professional judgement of the person administering or supervising the administration of the compositions.
  • Dosage ranges set forth herein are exemplary only and do not limit the dosage ranges that may be selected by medical practitioners.
  • the amount of active compound in the composition may vary according to factors such as the disease state, age, sex, and weight of the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • an immunogenically effective amount of a compound of the invention can be provided, alone or in combination with other compounds, with an immunological adjuvant, for example, Freund's incomplete adjuvant, NSA3, dimethyldioctadecylammonium hydroxide, or aluminum hydroxide.
  • an immunological adjuvant for example, Freund's incomplete adjuvant, NSA3, dimethyldioctadecylammonium hydroxide, or aluminum hydroxide.
  • the compound may also be linked with a carrier molecule, such as bovine serum albumin or keyhole limpet hemocyanin to enhance immunogenicity.
  • Toxicity of the compounds of the invention can be determined using standard techniques, for example, by testing in cell cultures or experimental animals and determining the therapeutic index, i.e., the ratio between the LD50 (the dose lethal to 50% of the population) and the LD100 (the dose lethal to 100% of the population). In some circumstances however, such as in severe disease conditions, it may be necessary to administer substantial excesses of the compositions.
  • C6 glioma cells obtained from American Type Culture Collection, were grown in DMEM supplemented with 10% (v/v) fetal bovine serum (FBS), 10 ⁇ g/ml streptomycin and 10 units/ml penicillin. Twenty-four hours prior to and throughout, all cells were maintained in serum-reduced media (1% FBS). Where noted, cells were exposed to the following agents: vehicle (phosphate buffered saline, PBS), CNTF (20 ng/ml), CNTFR ⁇ (200 ng/ml) or Complex (CNTF + CNTFR ⁇ ).
  • vehicle phosphate buffered saline, PBS
  • CNTF ng/ml
  • CNTFR ⁇ 200 ng/ml
  • Complex CNTF + CNTFR ⁇
  • RT-PCR Reverse transcription- polymerase chain reaction
  • the cDNA product (1 ⁇ l) was mixed with Tris-HCl (pH 8.4; 20 mM), MgCl 2 (1.5 mM), dNTP (200 ⁇ M), Platinum Taq (2 U), and one set of oligonucleotide primers (200 ⁇ M) given in final concentrations in a final volume of 40 ⁇ l.
  • Samples were denatured for 5 min at 95 °C and then amplified for 30 cycles of 94°C/45 sec, 58°C/1 min, and 72°C/1 min. Twenty microlitres of each PCR sample was run on a 1.8 % agarose gel in parallel with a 1.5 kb DNA standard (Invitrogen Corp., Burlington, ON).
  • Cells were grown to 80% confluence and subsequently treated with agents every 24 h (with fresh media changes) for three days. Cells were then washed twice with PBS and fixed for 10 minutes with 70% ethanol containing 0.15 M NaCl. Following blockage of non-specific antibody binding using 10% normal goat serum in PBS for 1 h, cells were incubated with rabbit polyclonal anti-Cx43 antibody (1 :400 dilution, Sigma- Aldrich, Oakville, ON) for 1 h, rinsed with PBS and subsequently incubated with Alexa-Fluor-conjugated goat anti-rabbit IgG secondary antibody (1 :500 dilution, Molecular Probes, Eugene, OR).
  • C6 cells were treated with agents every 24 h (with fresh media changes) for three days, rinsed twice with PBS and scraped off the plates in lysis buffer (RIP A buffer supplemented with protease inhibitors (complete, Mini; Roche, Indianapolis, IN )). DNA in the lysate was sheared using a 22 gauge needle. Total cell lysate was collected following microcentrifugation at 10,000xg for 10 minutes and total protein concentration was determined using the BCA Protein Assay Kit (Pierce-BioLynx, Brockville, ON).
  • the membrane was incubated in anti-Cx43 antibody (1:400 dilution, Sigma- Aldrich) for 1 h, rinsed again with PBS, and then bathed in secondary antibody tagged with horseradish peroxidase (1 :20,000 dilution, CedarLane Lab. Ltd., Hornby, ON). Following three PBS rinses, the membrane was incubated in Supersignal (Pierce-BioLynx) and exposed to X-ray film to visualize antibody binding. To normalize protein loading the membranes were gently stripped of antibodies and immunoblotted for GAPDH (1: 20,000 dilution, CedarLane Lab. Ltd.).
  • Pre-Loading Gap junctional coupling of C6 cells was determined by the pre-loading method as described by Goldberg et al. (1995). Briefly, C6 cells were grown to confluence in 12-well plates. Twenty-four h prior to pre-loading, a media change containing the various agents was performed. The agents were present in all solutions throughout the procedures. Donor C6 cells were preloaded with dye solution (5 ⁇ M calcein-AM (Molecular Probes) and 10 ⁇ M Dil (Sigma- Alderich) in an isotonic (0.3 M) glucose solution) for 20 min in a humidified incubator (37°C, 5% CO 2 /95% air).
  • dye solution 5 ⁇ M calcein-AM (Molecular Probes) and 10 ⁇ M Dil (Sigma- Alderich) in an isotonic (0.3 M) glucose solution
  • donor cells were rinsed twice with glucose solution, trypsinized, suspended in growth media, and seeded onto recipient (unlabeled) cells at 1:500 ratio. After being maintained in the incubator for 3 h, cells were examined with a photomicroscope. Gap junctional communication was assessed by the passage of calcein from donor cells to the underlying recipient cells. Only cells that were coupled to at least one other cell were examined. Growth Curves
  • C6 cells were seeded into 12-well plates at 2 x 10 4 cells/well (designated Day -1). Cells received fresh media containing the various agents every 24 h commencing on Day 0. The number of cells in each well were counted on Days 0-4, 6 and 8 using a haemocytometer and trypan blue as a dilutant.
  • Results were expressed as means ⁇ standard error of the means of four or more independent procedures. Statistical comparisons were performed using one way analysis of variance with a P value of ⁇ 0.05 considered significant.
  • RT-PCR was performed on total RNA. Compared to GAPDH levels, a low amount of CNTF mRNA was present in C6 cells while CNTFR ⁇ mRNA could not be detected. Specificity of the CNTFR ⁇ primers was confirmed on adult mouse brain samples.
  • C6 cell cultures contain a mixed population of cells in regards to protein expression (Roser et al, 1991).
  • a heterogenous population of Cx43 -expressing cells was identified; some cells showed low Cx43 staining (-70% of the population) and others with very low staining (the remaining 30% of the population). While no detectable changes in the ratios of this heterogenous population were induced by CNTF, CNTFR ⁇ or Complex, immunocytochemical analysis revealed that Complex induced an increase in Cx43 compared to vehicle, CNTF and CNTFR ⁇ alone. This increase in Cx43 was detected in both the cytoplasm and at the cell membrane.
  • the C6 glioma cell line expresses the transcript for CNTF but not CNTFR ⁇ .
  • CNTF is administered in the presence of the soluble CNTFR ⁇ , an increase in Cx43 and gap junctional communication is detected in C6 cells, accompanied by a significant decrease in C6 cell growth rate.
  • Positive identification of CNTF mRNA in C6 glioma cells shows that this glioma is astrocytic in origin.
  • examination for CNTF provides an alternative method in determining whether a neoplastic cell has an astrocytic lineage.
  • This identification technique is highly useful, since the vast majority of astrocytomas, including both cell lines and in vivo biopsies, lack glial fibrillary acidic protein expression, a marker commonly used to identify and distinguish astrocytes from other CNS cell types (Deck et al, 1978).
  • C6 glioma cells are not immortalized reactive astrocytes. Although C6 cells lack CNTFR ⁇ , they respond to CNTF if co-administered with CNTFR ⁇ .
  • Cx43 in C6 cells induced by Complex shows that the CNTF signaling pathway can overcome the Cx43 expression deficiency in these cells.
  • Cells were plated onto 60 mm dishes (3 ml cell suspension) or 100 mm dishes (10 ml cell suspension) and maintained in a humidified incubator at 37°C in 95% air/5% CO 2 . Media was changed every 3 days thereafter in addition to shaking the cultures.
  • Astrocytes were treated with either vehicle (PBS), C ⁇ TF (20 ng/ml; R&D Systems), soluble C ⁇ TFR ⁇ (200 ng/ml; R&D Systems), or Complex (20 ng/ml C ⁇ TF + 200 ng/ml C ⁇ TFR ⁇ ).
  • vehicle PBS
  • C ⁇ TF 20 ng/ml; R&D Systems
  • soluble C ⁇ TFR ⁇ 200 ng/ml; R&D Systems
  • Complex 20 ng/ml C ⁇ TF + 200 ng/ml C ⁇ TFR ⁇ .
  • Cx43 protein cells were treated with agents in fresh Media I every 24 hours for a total of 3 days.
  • mR ⁇ A levels cells were treated with agents for 24 hours.
  • STAT3, ERK1 (p44 MAPK) or ERK2 (p42 MAPK) phosphorylation cells received media change and 24 hours later, agents were added for 5, 15 and 60 minutes.
  • the STAT3 and MAPK/ERK pathways were inhibited by administering the pharmacological agents AG490 (50 ⁇ M; Calbiochem)(Hodge et al, 2002) and U0126 (10 ⁇ M; Promega Corp.)(Favata et al, 1998), respectively, 45 minutes prior to adding vehicle or Complex.
  • Astrocyte monolayers were rinsed twice with PBS and harvested in radioimmuno- precipitation assay buffer (1% Nonidet P-40, 0.5% ⁇ sodium deoxycholate and 0.1% SDS in PBS) supplemented with protease inhibitors (mini,Complete; Roche) using a rubber policeman. The lysate was sheared using a 22 gauge needle and centrifuged at 10,000 xg for 10 minutes at 4°C. For Cx43 protein analyses, some samples were treated with alkaline phosphatase (calf- intestinal; Roche) to detect total Cx43 protein. Protein concentration of total cell lysate was determined using the BCA Protein Assay Kit (Pierce).
  • Protein samples 50 ⁇ g each
  • molecular weight markers Bio-Rad Lab.
  • SDS-polyacrylamide gel electrophoresis subsequently electro-transferred onto a nitrocellulose membrane for 1 hour at 80 watts.
  • the membrane was immunoblotted using appropriate primary antibodies (Cx43, Sigma; phospho-STAT3 and phospho-p44/42 MAPK, Cell Signaling Tech.) and subsequently incubated in secondary antibodies tagged with horseradish peroxidase (CedarLane Lab. Ltd.).
  • the blots were then incubated in Supersignal (Pierce) and exposed to Kodak X-Omat x-ray film to visualize antibody binding.
  • membranes were stripped of antibodies using 2- ⁇ mercaptoethanol (10 mM), SDS (2%), and Tris (62.5 mM, pH 6.7) for 30 min at 65°C and probed for glyceraldehyde-3 -phosphate dehydrogenase (GAPDH; CedarLane Lab. Ltd.), STAT3 (Cell Signaling Tech.), or ERK1 and ERK2 (Santa Cruz Biotechnology, Inc.).
  • GPDH glyceraldehyde-3 -phosphate dehydrogenase
  • STAT3 Cell Signaling Tech.
  • ERK1 and ERK2 Santa Cruz Biotechnology, Inc.
  • Scrape-Loading/Dye Transfer Assay Intercellular coupling between astrocytes was measured using a modified version of the scrape-loading technique described by el-Fouly et al. (1987). Briefly, media was aspirated from the cultures, cells were bathed in 50 ⁇ L of dye solution (0.1 % carboxyfluorescein and 0.1 % dextran tetramethylrhodamine (Molecular Probes) in PBS) and, subsequently, scraped using a surgical blade.
  • dye solution 0.1 % carboxyfluorescein and 0.1 % dextran tetramethylrhodamine (Molecular Probes) in PBS
  • Cytoplasmic R ⁇ A was isolated from astrocytes using the phenol-chloroform-isoamyl alcohol method outlined by Sambrook et al. (1989). Briefly, cells were washed with PBS and, subsequently, lysed using Lysis Solution (0.2 M ⁇ aCl, 20 mM MgCl 2 , and 20 mM Tris pH 8.8).
  • RT-PCR was performed on the R ⁇ A as for Example A. 3 ⁇ g of R ⁇ A was pretreated with D ⁇ Ase and subsequently reverse transcribed in a thermal cycler. For semiquantitative RT-PCR, 25 cycles were used to amplify the cD ⁇ A to avoid saturation. Primer sets used for amplification are provided in Table I. The amplified cD ⁇ A was run on a 1.8% agarose gel containing ethidium bromide
  • the blots were washed 30 min each with 2X SSC (40°C), IX SSC (40°C), and 0.1X SSC with 0.1% SDS (42°C) and then exposed to Kodak Bio-Max film at -80°C for 6-48 hours using intensifying screens.
  • CNTF and its receptor, CNTFR ⁇ cortical astrocytes matured in vi ⁇ ro was compared to that in the cerebral cortex of adult mice.
  • RT-PCR analysis revealed that astrocytes expressed the transcript for CNTF but did not transcribe mRNA for the receptor subunit CNTFR ⁇ .
  • both CNTF and CNTFR ⁇ were detected at their expected product sizes. No transcripts were observed when the same RNA samples were amplified in the absence of reverse transcriptase.
  • astrocytes matured in culture exhibit a CNTF/CNTFR ⁇ phenotype comparable to astrocytes in an uninjured or non-reactive state.
  • CNTF Complex Increases Cx43 Protein Expression and Intercellular Coupling
  • Cx43 protein Localization of the Cx43 protein was examined by immunocytochemistry following three-day treatment of the astrocytes with vehicle, CNTF, CNTFR ⁇ or Complex.
  • the protein distributed intracellularly as well as to the periphery of the cells.
  • no noticeable difference in either immunostaining intensity or localization of the Cx43 was detected in cells treated with either CNTF or CNTFR ⁇ .
  • astrocytes treated with Complex showed a dramatic increase in Cx43 immunolabeling. This Complex-induced increase in Cx43 could be detected both within the cytoplasm and at the periphery of the cells.
  • MAPK/ERK pathway and the JAK/STAT pathway were employed.
  • Astrocytes were treated with the ERK-activating kinase (MEK)l/2 inhibitor l,4-diamino-2,3-dicyano-l,4bis[2- aminophenylthio]butadiene (U0126) for 45 minutes prior to and throughout the addition of Complex.
  • Phosphorylation of the ERK1/2 normally induced by Complex was inhibited by U0126.
  • astrocytes were pretreated with the JAK2/JAK3 inhibitor ⁇ - cyano-(3,4-dihydroxy)-N-benzylcinnamide (AG490).
  • JAK3 expression is limited to hematopoietic and lymphoid cells (Kawamura et al, 1994; Rane and Reddy, 1994) and has not been shown to be associated with CNTF signaling (Stahl et al, 1994).
  • AG490 inhibited most, but not all, phosphorylation of STAT3 upon addition of Complex.
  • RT-PCR analysis for Cx43 mRNA was performed on astrocytes treated with vehicle or Complex in the presence or absence of the pathway inhibitors, Complex was still capable of inducing an increase in Cx43 mRNA levels in the presence of the inhibitor solvent (DMSO) and in the presence of U0126.
  • DMSO inhibitor solvent
  • the Complex-induced increase in Cx43 mRNA was not observed in cells pretreated with AG490.
  • both CNTF and Complex induce phosphorylation of STAT3 protein, only
  • astrocytes in adult brain or matured in culture express other connexins, including Cx30 (Dahl et al, 1996; Kunzelmann et al, 1999; Nagy et al, 1999).
  • Cx30 Dahl et al, 1996; Kunzelmann et al, 1999; Nagy et al, 1999.
  • mRNA isolated from astrocytes treated with vehicle, CNTF, CNTFR ⁇ , or Complex was analysed by RT-PCR. No change in Cx3 transcript levels could be detected following 24 hour treatment with the agents examined.
  • astrocytes cultured from neonatal mice were matured in vilro for seven weeks prior to the described procedures the described procedures. These astrocytes have a mature, non-reactive phenotype, including expression of Cx30 (Dahl et al, 1996; Nagy et al, 1999; Dermietzel et al, 2000; Ip et al, 1993a; Kordower et al, 1997; Maclennan et al, 1996; Monville et al, 2001; Squinto et al, 1990).
  • Previous studies demonstrated that astrocytes exhibit inconsistent or highly variable responses to CNTF. Different states of reactivity exist for astrocytes both in vivo and in vitro, from normal to reactive.
  • Reactive astrocytes are known to express both mRNA and protein for CNTFR ⁇ (Duberley et al, 1995; Duberley and Johnson, 1996; Ip et al, 1993b; Lee et al, 1997; Rudge et al, 1994a; Rudge et al, 1995). Astrocytes isolated and cultured from neonatal animals and subsequently maintained in culture for a long duration are in a non-reactive state.
  • Example mature murine cortical astrocytes are shown to express CNTF but not CNTFR ⁇ . Only when CNTF was administered in the presence of CNTFR ⁇ was upregulation of both Cx43 mRNA and protein observed. The regulation of Cx43 expression by Complex was dependent on the JAK/STAT pathway but not the MAPK/ERK pathway. The ability of Complex to upregulate Cx expression is selective as the expression of Cx30 is not affected. Although an increase in Cx43 was detected within the cytoplasm, Complex induced an increase in Cx43 mainly at the periphery of the cells.
  • Cx43 becomes phosphorylated, forms a connexon, which subsequently combines with the connexon from an adjacent cell to form the gap junction (reviewed by Lampe and Lau, 2000; Musil and Goodenough, 1991).
  • astrocytes are typically highly coupled, Complex further increased or stabilized functional intercellular communication via gap junction formation.
  • Complex also increases Cx43 and gap junctional coupling in C6 cells as shown in Example A. Therefore, upregulation of Cx43 and intercellular communication by Complex occurs in both gap junction-competent and -deficient cells of the CNS. Upregulation of Cx43 and gap junctions by Complex will circumvent extensive tissue damage in brain injuries, insults and in progressive CNS diseases.
  • CNTF has shown limited neuroprotective effects within the CNS following various insults (Kumon et al., 1996; Ogata et al., 1996; Unoki and LaVail, 1994; Wen et al., 1995b), the limitation now appears to be due to the lack of CNTFR ⁇ on astrocytes.
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • F12 fetal bovine serum
  • Neurobasal Medium penicillin-streptomyosin
  • HBSS Hank's balanced salt solution
  • PBS Phosphate Buffered Saline
  • Earle's Balanced Salt Solution were obtained from GIBCO Laboratories (Burlington, ON, Canada).
  • Cytosine arabinoside poly-D-lysine, D- glucose, insulin, transferrin, putrescine-HCl, progesterone, sodium selenite, CBX, GZA, AGA, propidium iodide, Hoechst 33342 dye, glutamate, anti-microtubual associated protein 2 (MAP2) antibody, anti-glial fibrillary acidic protein (GFAP) antibody and lactate dehydrogenase (LDH) Detection kit were products of Sigma (Oakville, ON, Canada). L- [ 3 H]glutamate ([ 3 H]Glu, 38-46 Ci/mmol) was purchased from Amersham Canada (Oakville, ON, Canada).
  • Apoptosis Detection System (terminal dUTP nick end labeling, TUNEL, Fluroescein) was from Promega (Madison, WI, USA). Cell strainers (pore size 70 ⁇ m) and 6- well plates were acquired from NWR (Mississauga, ON, Canada). Dil (l,l'-dioctadecyl- 3,3,3',3'-tetramethylindocarbocyaine perchlorate) and calcein acetoxymethyl ester (calcein- AM) were purchased from Molecular Probes (Eugene, OR, USA). Solution I consisted of HBSS supplemented with 10%> FBS.
  • Media I was comprised of Neurobasal Medium and DMEM/F12 (2:3) supplemented with 10% FBS.
  • Media II was prepared with 54 ml Neurobasal Medium, 36 ml DMEM/F12, 50 ⁇ l penicillin-streptomyosin, and 2 ml of N2 Supplements.
  • N2 Supplements (Mazzoni, I.E., et al, 1991) consisted of the following (given in final concentrations in Media II): glucose (0.6%), insulin (10 ⁇ g/ml), transferrin (20 ⁇ g/ml), putrescine-HCl (62 ⁇ M), progesterone (20 nM), and sodium selenite (30 nM).
  • Sodium Hepes Buffer contained (final concentrations) 134 mM NaCl, 5.2 mM KC1, 1.8 mM CaCl 2 , 0.8 mM MgSO 4 , 10 mM glucose, and 20 mM HEPES, pH 7.3.
  • astrocytes were prepared from murine cortices using the method described for Example B. Cortices were dissected from 1 to 2 day old mouse pups, placed into PBS, and subsequently freed of meninges. Cortices were then placed into DMEM supplemented with 10% FBS and were mechanically dissociated using a serological pipette. This cell suspension was then passed through a cell strainer and diluted using DMEM supplemented with 10% FBS and penicillin-streptomyosin at a ratio of 7 ml/cortex. Cells were plated onto 6-well plates (previously coated with poly-D-lysine, 100 ⁇ g/ml) in 2 ml aliquots.
  • Cultures were maintained within a humidifed incubator at 37°C in 95% air/5% CO 2 . Culture media was continuously replaced every three days in addition to shaking the cultures. Astrocytic cultures were maintained for 15-17 days from initial plating prior to use or co- culturing with neurons. Immunocytochemical characterization of cell types present in the final astrocytic cultures revealed that 95-97% of the population were GFAP immunopositive.
  • Neurons were prepared from mouse cortices in a modified method by Mazzoni and Kenigsberg (1991). Cortices were dissected from embryonic mice of gestation age 16 days and placed into HBSS supplemented with 10% FBS. Cortices were then freed of meninges, minced with scalpels and mechanically disrupted by gently triturating through a fire-polished pasteur pipette (bore size of approximately 0.6 mm internal diameter). Cell suspension was passed through a cell strainer, centrifuged, and subsequently resuspended in HBSS and Media 1 (1:1) supplemented with 10% FBS. Cells (1 x 10 6 cells/well) were seeded on top of astrocyte cultures and allowed to settle for 2 hours.
  • Co-cultures consisted of a monolayer of astrocytes to which cortical neurons were seeded on top.
  • Blocking of astrocytic gap junctions was determined by dye coupling using the preloading method as described by Goldberg et al. (1995). Astrocyte cultures were maintained in Media II for 7 days. Narious concentrations of the blocking agents CBX and AGA and the inactive analogue GZA were tested; all agents were added to media 1 hour prior to pre-loading and were present in all solutions.
  • Blocking agents were added to co-cultures 1 hour prior and were present in all solutions throughout the procedure. Co-cultures were rinsed with PBS and then bathed in EBSS with glutamate (1 mM) or sham (vehicle) for 3 hours in a humidified incubator (37°C, 95% air/5% CO 2 ). Cells were subsequently maintained for 24 hours post glutamate/sham exposure in fresh Media II.
  • Glutamate uptake procedures were performed in a manner similar to that described by Sitar et al. (1999). Confluent cultures of astrocytes were switched to Media II for 1 week prior to procedures. Blocking agents were added to cultures 1 hour prior to glutamate uptake and were present in all solutions during transport. Cells were rinsed with Sodium Hepes Buffer and exposed to glutamate solution for 120 seconds. Uptake was terminated by washing cultures with ice-cold Tris-sucrose (pH 7.3) solution. Cells were harvested by osmotic lysis (1 ml water/dish) and mechanical scraping. The radioactive contents of the buffer and cells were measured by liquid scintillation counting. Glutamate uptake was computed based on the specific activity of radiolabeled glutamate in the buffer and taken-up by the cells.
  • a minimal concentration of CBX and AGA required to block astrocytic gap junctions was determined by assaying unlabeled cells receiving calcein from a labeled cell (Dil and calcein). Final concentrations of 0 (vehicle), 1, 10, 25 and 50 ⁇ M of blockers were tested. While vehicle (water) and GZA had no affect on the passage of calcein from donor cells to, and among, recipient cells, concentrations including and exceeding 25 ⁇ M CBX effectively blocked the passage of this dye. A lower concentration of AGA (10 ⁇ M) was required to similarly block astrocytic gap junctions while its vehicle (DMSO) had no effect. From the results obtained in pre-loading, concentrations of 25 ⁇ M CBX and 10 ⁇ M AGA were chosen for use in the remainder of this Example.
  • Glutamate cytotoxicity was analyzed by three mechanisms: LDH release, the inability to exclude propidium iodide, and TUNEL labeling.
  • LDH release When presented with a sham insult, neither vehicle, CBX nor GZA caused the cells to release a significant amount of LDH.
  • glutamate was administered in the presence of vehicle or GZA, no significant LDH release occurred.
  • gap junctions were blocked with CBX, a similar glutamate insult caused the cells to release a significant amount of LDH.
  • Gap Junction Blocker Does Not Affect Astrocytic Glutamate Transporters
  • This Example demonstrates the important role of intercellular communication that allows cells to endure high levels of extracellular glutamate.
  • Glutamate can induce a cytotoxic effect to sensitive cells (i.e. neurons) by one of two mechanisms.
  • glutamate activates ionotrophic receptors which subsequently depolarizes the cell resulting in cell swelling and, potentially, necrosis.
  • glutamate metabotrophic receptor overstimulation may cause the cell to undergo transcriptionally active suicide (apoptosis).
  • apoptosis transcriptionally active suicide
  • PI and TUNEL staining indicated that the dosage of glutamate employed significantly insulted the cells. More specifically, the PI and TUNEL labelling was highly associated with the glutamate-sensitive neurons and labelled very few underlying astrocytes (similar results to Choi (1987)). Astrocytic tolerance to the glutamate insult is likely due to astrocytes lacking NMDA receptors and housing large glycogen stores (a crucial metabolite for energy production).
  • the increased glutamate cytotoxicity in the presence of the gap junction blockers seems directly related to the blockers impeding intercellular gap junctional communication.
  • the blockers alone caused no significant cell death and no significant difference in mortality was determined between vehicle and the inactive blocker in the presence of glutamate.
  • the cell suspension was passed through a cell strainer, centrifuged at 5000 xg for 5 minutes, and resuspended in Hank's Balanced Salt Solution (Invitrogen Corp.) and Media I (Neurobasal Medium and DMEM/F12 (2:3); Invitrogen Corp.) in a 1 :1 ratio supplemented with 10% FBS.
  • Immunocytochemistry Co-cultures were rinsed with phosphate buffered saline (PBS) and fixed with 4% formaldehyde (in PBS) for 10 minutes. Non-specific binding of antibodies was blocked by incubating the cultures with 10% normal goat serum (in PBS). Cultures were then incubated with microtubule associated protein 2 (MAP2) monoclonal antibody and glial fibrillary acidic protein (GFAP) polyclonal antibody (Sigma). Following three washes with PBS, cultures were incubated in Alexa- and fluorescein-conjugated secondary IgG (Molecular Probes) for 1 hour and then mounted with Nectashield mounting medium (Nector). Immunostaining was viewed using a Zeiss Axioskop microscope and images were captured using Northern Exposure, Nersion2 (ImageExperts Inc.).
  • MAP2 microtubule associated protein 2
  • GFAP glial fibrillary acidic protein
  • Co-cultures were rinsed with PBS and then bathed in Earle's Balanced Salt Solution (Invitrogen Corp.) with glutamate (1 mM) or PBS (sham insult) for 3 hours in a humidified incubator (37°C, 95% air/5% CO 2 ). Following the insult, cells were maintained in fresh Media II.
  • Earle's Balanced Salt Solution Invitrogen Corp.
  • glutamate 1 mM
  • PBS sham insult
  • the stained cells were rinsed three times with PBS and mounted using Nectashield mounting medium (Nector). Staining was viewed and images were captured as outlined above.
  • TUNEL-labelling vehicle pretreatment
  • Similar glutamate-induced cytotoxicity as marked by TUNEL-labelling was seen in CNTFR ⁇ -pretreated co-cultures.
  • considerably less number of cells stained with TUNEL was observed in the co-cultures pretreated with CNTF and Complex. Comparisons between groups revealed that only Complex pretreatment significantly reduced the number of TUNEL-positive cells, as compared to vehicle pretreatment.
  • CNTF Glutamate-induced cytotoxicity in neuron and astrocyte co-cultures is reduced by CNTF pretreatment. Since neurons express CNTFR ⁇ , CNTF alone can directly activate those cells and potentially increase their resilience to glutamate excitotoxicity. Skaper et al. (1992) but the extended neuroprotection shown in this Example requires administration of both CNTF and CNTFR ⁇ .
  • CNTF causes a reduction in glutamate-induced cytotoxicity within its own pretreatment group, this difference is not significantly different from vehicle pretreatment under similar conditions.
  • CTNF CTNF in combination with CNTFR ⁇
  • the "Complex" of CNTF-(soluble)CNTFR ⁇ was administered to mice following induced stroke. Stroke was induced in mice by middle cerebral artery occlusion (MCAO). Occlusion of the blood vessel in mice is a widely used stroke model (Siushansian et al., 2001 ; Nakase et al., 2003). Immediately following the insult (2 hour occlusion in the mouse model), the respective agents (vehicle, CNTF, or Complex) were administered via intraperitoneal injection. Since CNTF administration into both rodents and humans induces weight reduction, the body weights and food intake (provided ad libum) were closely monitored.
  • MCAO middle cerebral artery occlusion
  • infarct core and penumbra
  • TUNEL terminal dUTP nick end labeling
  • a large portion of the body weight reduction can be attributed to the agents inducing loss of both appetite and thirst since food and H 2 O intake were significantly reduced compared to vehicle.
  • the loss in appetite is solely attributed to the effect of Complex and independent of the stroke since Complex injection without stroke caused similar results to that of Complex with insult.
  • the initial concentration of Complex chosen for injections to reduce stroke-induced brain damage was that in which the same concentration of CNTF alone would induce physiological changes (i.e. weight loss; Fantuzzi et al., 1995).
  • concentration of Complex may be titered to determine the amount most effective for neuroprotection in vivo, by for example reducing brain tissue damage without causing substantial side-effects.
  • MCAO insults are performed on knockout mice. While CNTFR ⁇ knockout mice are not viable, mice lacking CNTF expression thrive normally. Using mice in which the CNTF gene has been deleted through homologous recombination (generous gift of Dr. Micheal Sendtner, Universitaat W ⁇ urzburg), the importance of CNTF expression in circumventing brain injury following stroke is assessed. A similar MCAO insult and similar methods to assess brain injury as outlined herein is performed on the CNTF knockout mice and compared to wild-type littermates. Furthermore, in cases where the lack of endogenous CNTF expression leads to increased brain injury in the knockout mice, rescue of brain injury by administration of exogenous CNTF or Complex following the insult is determined.
  • RNA RNA which will be analyzed by, for example, gene arrays.
  • Narious genes are differentially expressed by treatment of cultures with Complex compared to vehicle or C ⁇ TF treatment.
  • the identified differential gene candidates are confirmed by other techniques (e.g. northern blot analysis and/or reverse transcription polymerase chain reactions).
  • Connexin43 a protein from rat heart homologous to a gap junction protein from liver. J Cell Biol 105:2621-2629. Blanc EM, Bruce-Keller AJ, and Mattson MP (1998) Astrocytic gap junctional communication decreases neuronal vulnerability to oxidative stress-induced disruption of Ca2+ homeostasis and cell death. J Neurochem 70:958-970. Blottner D, Bruggemann W, and Unsicker K (1989) Ciliary neurotrophic factor supports target- deprived preganglionic sympathetic spinal cord neurons. Neurosci Lett 105:316-320. Brosnan CF, Scemes E, and Spray DC (2001) Cytokine regulation of gap junction connectivity: an open-and-shut case or changing partners at the Nexus? Am J Paihol
  • GD (1991) The receptor for ciliary neurotrophic factor. Science 253:59-63. Davis, S., T.H. Aldrich, N. Stahl, L. Pan, T. Taga, T. Kishimoto, N.Y. Ip, and G.D.
  • Ciliary neurotrophic factor induces serum amyloid A, hypoglycaemia and anorexia, and potentiates IL-1 induced corticosterone and IL-6 production in mice.
  • TUNEL assay In situ detection of fragmented DNA (TUNEL assay) fails to discriminate among apoptosis, necrosis, and autolytic cell death: a cautionary note.
  • TGF-beta 1 protects hippocampal neurons against degeneration caused by transient global ischemia. Dose-response relationship and potential neuroprotective mechanisms. Stroke 27:1609-1614. Hertz L. Possible role of neuroglia: a potassium-mediated neuronal—neuroglial—neuronal impulse transmission system. Nature 1965;206:1091-1094 Hirano T, Nakajima K, and Hibi M (1997) Signaling mechanisms through gpl30: a model of the cytokine system. Cytokine Growth Factor Rev 8:241-252. Hodge, D.R., D. Li, S.M. Qi, and W.L. Farrar. 2002. IL-6 induces expression of the Fli-1 proto-oncogene via STAT3. Biochem. Biophys. Res. Commun. 292:287-291.
  • JNeurosci 11:3124-3134 Ip NY, McClain J, Barrezueta NX, Aldrich TH, Pan L, Li Y, Wiegand SJ, Friedman B, Davis S, and Yancopoulos GD (1993a) The alpha component of the CNTF receptor is required for signaling and defines potential CNTF targets in the adult and during development. Neuron 10:89-102. Ip NY, Wiegand SJ, Morse J, and Rudge JS (1993) Injury-induced regulation of ciliary neurotrophic factor mRNA in the adult rat brain. Eur JNeurosci 5:25-33. Ip, N.Y. and G.D. Yancopoulos. 1992. Ciliary neurotrophic factor and its receptor complex.
  • Kandler K Katz LC. Neuronal coupling and uncoupling in the developing nervous system.
  • C ⁇ TF ciliary neurotrophic factor
  • Ciliary neurotrophic factor may activate mature astrocytes via binding with the leukemia inhibitory factor receptor.
  • Nedergaard M (1994) Direct signaling from astrocytes to neurons in cultures of mammalian brain cells. Science 263:1768-1771.
  • Nedergaard M Cooper AJ, Goldman SA. Gap junctions are required for the propagation of spreading depression. J Neurobiol 1995;28:433-444 Negro, A., G. Corona, E. Bigon, I. Martini, C Grandi, S.D. Skaper, and L. Callegaro. 1991.
  • JAK3 a novel JAK kinase associated with terminal differentiation of hematopoietic cells.
  • Fibroblast growth factors-5 and -9 distinctly regulate expression and function of the gap junction protein connexin43 in cultured astroglial cells from different brain regions. Glia 30:231-241. Reuss, B., R. Dermietzel, and K. Unsicker. 1998. Fibroblast growth factor 2 (FGF-2) differentially regulates connexin (ex) 43 expression and function in astroglial cells from distinct brain regions. Glia 22:19-30. Robe PA, Rogister B, Merville MP, and Bours V (2000) Growth regulation of astrocytes and
  • RNA from eukaryotic cells in Molecular Cloning: A Labora ⁇ ory Manual Cold Springs
  • Acidic fibroblast growth factor prevents death of hippocampal CA1 pyramidal cells following ischemia.
  • Gap junctions modulate survival-promoting effects of fibroblast growth factor-2 on cultured midbrain dopaminergic neurons.
  • Tanaka K Tanaka K
  • Tsukahara T Hashimoto N
  • Ogata N Hashimoto N
  • Yonekawa Y Kimura T
  • Taniguchi T (1994) The role of brain-derived neurotrophic factor in transient forebrain ischemia in the rat brain. Neurosurgery 34:323-331. Tsuzuki N, Miyazawa T, Matsumoto K, Nakamura T, and Shima K (2001) Hepatocyte growth factor reduces the infarct volume after transient focal cerebral ischemia in rats. Neurol
  • Zahs KR Heterotypic coupling between glial cells of the mammalian central nervous system.
  • Accession numbers refer to Accession numbers from multiple databases, including GenBank, the European Molecular Biology Laboratory (EMBL), the DNA Database of Japan (DDBJ), or the Genome Sequence Data Base (GSDB), for nucleotide sequences, and including the Protein Information Resource (PIR), SWISSPROT, Protein Research Foundation (PRF), and Protein Data Bank (PDB) (sequences from solved structures), as well as from translations from annotated coding regions from nucleotide sequences in GenBank, EMBL, DDBJ, or RefSeq, for polypeptide sequences. Numeric ranges are inclusive of the numbers defining the range.

Abstract

The invention provides, methods for neuroprotection using a CNTF peptide and a peptide that acts as a receptor for CNTF, e.g., CNTFR-alpha. The combination of the invention is more effective than that achieved in the absence of treatment (i.e., without applying exogenous agents) or by treatment with CNTF alone, or with CNTF receptor alone.

Description

METHODS FOR NEUROPROTECTTON
FIELD OF THE INVENTION
The invention relates, in part, to methods for neuroprotection.
BACKGROUND OF THE INVENTION
Acute and chronic neurodegeneration in the central nervous system (CNS), for example, as a result of trauma, stroke, ischemia, Huntington's disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis (ALS), olivopontocerebellar atrophy, epilepsy and hypoglycemic encephalopathy, is believed to result from a general process termed "excitotoxicity.," which is mediated by the action of glutamate.
Glutamate is a negatively charged amino acid used for excitatory synaptic transmission in the mammalian nervous system. Although the concentration of glutamate can reach the millimolar range in nerve terminals (Waelsch, H., 1951), its extracellular concentration is maintained at a low level to prevent neurotoxicity. As early as 1957, Lucas and Newhouse
(1957) noted that glutamate can be toxic to neurons if presented at a high concentration. Later, Olney and Sharpe (1969) implemented the term "excitotoxicity" to describe the cytotoxic effect that glutamate (and other such excitatory amino acids) has on neurons when applied at high dosages. Physiologically, high levels of glutamate can be achieved by excessive release, inhibition of uptake, or both. Normally, a low concentration of extracellular glutamate is maintained by both neurons and astrocytes. Neurons store glutamate in intracellular stores and regulate its release (Reagan, R.F., 1994). Astrocytes take up extracellular glutamate by specific transporters and convert the glutamate into glutamine which is then released for neuronal t uptake [Bergles, D.E. (1997); Robinson, M.B. (1997); Rothstein, J.D. (1996); Tanaka (1997)]. ' Thus, in the process of excitotoxicity, glutamate is released in a self-perpetuating manner by the neurons, resulting in excessive or prolonged activation of glutamate receptors.
The conjunction of such excessive glutamate stimulation on the energy-depleted neurons taken with the compromised ability of the neurosupportive astrocytes to sequester toxic levels of extracellular glutamate leads to neuronal death via necrosis and apoptosis. Narious interventions are currently being examined to reduce neuronal death associated with central nervous system injuries and diseases (Kermer, 1999). Such therapies include glutamate release . _ inhibitors, glutamate receptor antagonists, Ca2+ channel blockers, GABA receptor agonists, gangliosides, neurotrophic factors, calpain inhibitors, caspase inhibitors, free radical scavengers, irnmuno- and cell metabolism modulators. Neuroprotection during brain injuries and within the central nervous system (CNS) may be linked to the presence of gap junctions and their constitutive proteins (Ozog et al, 2002b; Naus et al., 2001; Ozog et al, 2002a; Siushansian et al, 2001; Iwata et al, 1998; Nishimune et al, 2000; Enkvist and McCarthy, 1994; Cotrina et al, 1998). Gap junctions are proteineous channels that directly link the cytosol of adjacent cells and allow intercellular passage of molecules of Mτ <1200 (Kumar and Gilula, 1996). The structural unit of the gap junction is the connexon which itself consists of protein subunits termed connexins (Cxs)(Kumar and Gilula, 1996). At least eight different Cxs have been identified in the CNS. The connexins (Cxs) are designated according to relative molecular mass and numerous isoforms have been identified, including at least eight within the CNS (Dermietzel and Spray, 1993; Rozental et al., 2000; Willecke et al, 2002). Cx43, the major Cxn expressed by astrocytes, has also been reported to be in neurons (Dermietzel and Spray, 1993; Nadarajah et al., 1996; Alvarez-Maubecin et al, 2000; Nedergaard, 1994; Micevych and Abelson, 1991; Micevych et al, 1996). Astrocytes play a supportive role for neurons by providing crucial metabolites, maintaining favourable extracellular levels of neurotransmitters and ions, and helping constitute the blood-brain barrier (Hatten and Mason, 1986). Unlike neurons, astrocytes also have the ability to readily divide. Normal astrocytes are highly coupled by gap junctions and express primarily Cx43 and to a lesser extent Cxs 26, 30, 31.1 and 40 (Dermietzel et al, 2000).
The expression and activity of gap junctions can be altered by short-term and long-term regulation (reviewed by Giaume and McCarthy, 1996; Rouach and Giaume, 2001). Short-term regulation occurs within milliseconds to minutes and is the result of a change in the biophysical properties of the channel. Such channel gating is usually caused by post-translational processing such as phosphorylation or channel blocking. Long-term regulation, however, occurs over hours or days and consists of the formation or disappearance of gap junction channels as a result of changes to gene transcription and post-translational processes. While numerous agents have been identified to modulate short-term gap junction regulation (reviewed by Dhein, 1998), long-term regulators have not been extensively studied.
Gap junctional coupling can be impaired/blocked by administering various agents including carbenoxolone (3 -hydroxy- l l-oxoolean-12-en-30-oicacid 3 hemisuccinate; CBX) and 18α-glycyrrhetinic acid (3-hydroxy-ll-oxo-18,20-olean-12-en-29-oic acid; AGA). CBX is a water-soluble synthetic gap junction blocker and has an inactive analog, glycyrrhizic acid (GZA), which is a derivative of Glycyrrhiza glabra. AGA is also derived from C glabra and also blocks gap junctions; the exact mechanism to which AGA and CBX block the junctions has not yet been deciphered. Previous studies have reported that these blocking agents have no effect on cell viability if presented at concentrations less than 100 μM.
In the CNS, Cx43 is upregulated in astrocytes treated with transforming growth factor- βl (Robe et al, 2000) but downregulated by both interleukin (IL)-lβ (John et al, 1999) and transforming growth factor-β3 (Reuss et al., 1998). Fibroblast growth factor-2 has opposing effects on cells of the CNS, increasing Cx43 expression in neurons (SiuYi et al, 2001) but decreasing this gap junction component in astrocytes (Reuss et al., 1998; Reuss et al, 2000). Prior research suggests that Cx43 is up-regulated following a CNS insult (Enkvist and McCarthy, 1994; Hossain et al, 1994). In regards to neuroprotection in brain injury and disease, the specific role of gap junctions is not yet clear. Gap junctions could have a direct protective effect, allowing the astrocytic syncytium to buffer extracellular space from cytotoxic levels of metabolites and ions (such as glutamate and K+). In addition, gap junctions could promote the viability of energy- exhausted cells by allowing essential molecules (e.g. ATP and glucose) to move into areas of high demand. Alternatively, acting in a destructive manner, gap junctions could permit the movement of potentially toxic metabolites into cells that are already compromised by the insult.
Ciliary neurotrophic factor (CNTF) is a cytokine normally found within the cytosol of astrocytes. CNTF was originally identified, purified and cloned based upon its ability to support the in viiro survival of parasympathetic neurons (Barbin et al, 1984; Lin et al, 1989; Masiakowski et al., 1991; Stockli et al, 1989). CNTF is a member of the IL-6 family that includes IL-6, IL-11, oncostatin M, cardiotrophin-1 and leukemia inhibitory factor (LIF)(Ip and Yancopoulos, 1992; Lin et al, 1989; Patterson, 1992; Stockli et al, 1989); all of these family members share the signaling receptor system of glycoprotein 130 (gpl30) and LIF receptor-β (LIFRβ) to initiate intracellular signaling cascades. The expression of CNTF is almost exclusively restricted to nervous tissue, where it is normally produced by astrocytes in the CNS and Schwann cells in the peripheral nervous system (Stockli et al, 1991).
CNTF is considered a "brain injury" cytokine based on the findings that: 1) astrocytes express and retain CNTF as a cytosolic protein which is only released following brain disturbances (Stockli et al, 1989; Stockli et al, 1991; Rudge et al., 1994a; Rudge et al.,
1994b); 2) the CNTF specific receptor, CNTFR-alpha or CNTFRα, is located on neurons (Ip et al, 1993a; Kahn et al, 1997; Maclennan et al, 1996) and post-insulted (reactive) astrocytes (Ip et al., 1993b; Rudge et al, 1995); and 3) expression of both CNTF and CNTFRα is upregulated following disturbances to the CNS (Asada et al., 1995; Asada et al, 1996; Duberley et al, 1995; Ip et al, 1993b; Lee et al, 1997; Lin et al, 1998; Nieto-Sampedro et al, 1982; Park et al, 2000; Rudge et al, 1994a; Wen et al, 1995a). Upon release by injured astrocytes, CNTF binds to CNTFRα and subsequently recruits gpl30 and LIFRβ through which a cascade of several signal transduction pathways is initiated. CNTFRα can be cleaved (i.e., by PLC) from expressing cells and, as a soluble form, can bind CNTF and activate cellular pathways.
Upon its release, CNTF may bind its receptor (CNTFRα; soluble or membrane bound form) and subsequently activate a cascade of signal transduction pathways. The predominant pathways activated by CNTF are the mitogen-activated protein kinase/extracellular signal- regulated kinase (MAPK/ERK) pathway and the j anus kinases/signal transducers of activated transcription (JAK/STAT) pathway (reviewed by Monville et al, 2001). Both MAPK/ERK and JAK/STAT pathways lead to direct transcriptional activation of various genes (Symes et al, 1994). CNTFRα lacks τransmembrane and cytoplasmic domains and is instead anchored to the cell surface by a glycosyl-phosphotidylinositol linkage (Davis, et al. 1991). This linkage can be cleaved by phospholipases and release the receptor as a soluble protein. Although the soluble receptor is functional, it requires CNTF to be released into the extracellular space by dying or compromised cells to which it can bind and subsequently activate the ubiquitously expressed β components of the receptor. Davis, et al. (1993b) demonstrated that administration of both the soluble receptor and CNTF to various hematopoietic cell lines mimicked the effect of leukemia inhibitory factor (LIF) on these cells. The hematopoietic cell lines were not normally responsive to CNTF alone and did not express the receptor. However, only hematopoietic cell lines expressing gpl30 and the β receptor for LIF responded to the combination of CNTF and its soluble receptor. The authors speculated that CNTFRα may participate in interactions between the nervous system and hematopoietic and other systems. Similarly, combination of the receptor and CNTF has been shown to promote macrophage chemotaxis in a manner similar to interleukin 6 (Kobayashi, H. & Mizisin, A.P., 2000).
SUMMARY OF THE INVENTION
This invention arises from the discovery that co-administration of a CNTF peptide and at a peptide that acts as a receptor for CNTF provides a neuroprotective effect on CNS tissue exceeding the neuroprotective effect of CNTF alone. These effects appear to be brought about by the action of a combination of the receptor and CNTF on mature or un-reactive astrocytes, or alternatively, neoplastic cells of astrocytic origin.
Neuroprotection provided by this invention includes protection from damage resulting from neural injury or insult and from neurotoxicity, including excitotoxicity. Thus, neuroprotection provided by this invention will be useful in the treatment of acute and chronic neurodegenerative disorders that may involve excitotoxicity, for example glutamate excitotoxicity, including stroke/ischemia, trauma, epilepsy, Huntington's Disease, amyotrophic lateral sclerosis and hypoglycemic encephalopathy. Neuroprotection provided by this invention may be brought about upon injured or diseased tissue or in a preventative fashion during or prior to events expected to lead to a neural insult.
The invention provides methods for providing neuroprotection; for inhibiting cell degeneration or cell death; for treatment or prophylaxis of a neurodegenerative disease; or for ameliorating the cytotoxic effect of a compound (for example, a excitatory amino acid such as glutamate; a toxin; or a prophylactic or therapeutic compound that exerts a cytotoxic side effect) in a subject in need thereof, by administering to the subject an effective amount of a CNTF peptide or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding the CNTF peptide or a biologically-active fragment or variant thereof; and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof. In various embodiments, the methods of the invention include protection against excitotoxicity, for example glutamate excitotoxicity.
In various embodiments, the subject, for example, a human, may be suffering from neural insult or injury; or may be suffering from a condition selected from substance abuse, trauma, stroke, ischemia, Huntington's disease, Alzheimer's disease, Parkinson's disease, prion disease, variant Creutzfeld- Jakob disease, amyotrophic or hypoglycemic encephalopathy; or may be undergoing surgery or other intervention. The subject may have a pre-existing condition which would benefit by neuroprotection or the patient may be treated to reduce deleterious effects of a concomitant or subsequent neural injury, such as may occur during surgery or other intervention. In various embodiments, the peptide that acts as a receptor for CNTF is soluble (e.g., soluble CNTFRα). In various embodiments, the peptides are administered as a complex.
The invention also provides the use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof for neuroprotection; for modulation of cell degeneration or cell death; for treatment or prophylaxis of a neurodegenerative disorder; or for ameliorating the cytotoxic effect of a compound.
The invention also provides the use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof for preparation of a medicament for neuroprotection; modulation of cell degeneration or cell death; treatment or prophylaxis of a neurodegenerative disorder; or ameliorating the cytotoxic effect of a compound.
The invention also provides compositions including a CNTF peptide or a biologically- active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof for neuroprotection; modulation of cell degeneration or cell death; treatment or prophylaxis of a neurodegenerative disorder; or ameliorating the cytotoxic effect of a compound.
In various embodiments, the uses of the invention include protection against excitotoxicity, for example glutamate excitotoxicity.
The invention provides a method of screening for a neuroprotective compound, by providing a first system including CNTF or a biologically-active fragment or variant thereof; providing a second system including a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof; providing a third system including CNTF or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof; contacting the first and second systems with a test compound; and determining whether the test compound modulates neuroprotection in the first or second system when compared to the third system.
The invention also provides a method of screening for a neuroprotective compound, by providing a system including CNTF or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof; contacting the system with a test compound; and determining whether the test compound modulates neuroprotection in the presence or absence of a gap junction blocker.
The invention also provides a method of screening for a nucleic acid molecule that modulates neuroprotection, by providing a neuronal or neuronal-associated cell; contacting the neuronal or neuronal-associated cell with CNTF or a biologically-active fragment or variant thereof; a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof; or a combination comprising the CNTF and the peptide that acts as a receptor for CNTF; and determining the level of nucleic acid molecule expression (for example, by a microarray), where a change in the level of nucleic acid molecule expression in the combination, when compared to the level of nucleic acid molecule expression in either CNTF or the CNTF receptor systems alone, is indicative of a nucleic acid molecule that modulates neuroprotection.
In various embodiments, the neuroprotection may include protection of neuronal cells or neuronal-associated cells from injury or degeneration or increasing the expression level of a connexin (e.g. Cx43). The system(s) may include neuronal or neuronal associated cells. In a separate aspect, this invention also provides a method for treatment of a CNS tumor in a patient in need thereof, comprising administering to the patient an effective amount of the aforementioned complex or a pharmaceutical composition comprising the aforementioned complex. This separate aspect also provides the use of a complex of a peptide having the activity of CNTF and a peptide having the function of a receptor for CNTF the treatment of neoplastic disorders. This separate aspect also provides the use of the aforementioned complex for preparation of medicaments for treatment of neoplastic disorders. This anti-neoplastic aspect of the invention is distinct from the neuroprotection aspect.
The separate aspect of this invention also provides a method for determining the susceptibility of a neoplastic cell to suppression by the aforementioned complex, comprising determining whether the cell expresses CNTF. This method may further comprise determining whether the cell expresses CNTFRα. The method may also further comprise isolating and culturing neoplastic cells to be tested, and may further comprise obtaining a sample of tissue from a subject comprising such cells to be tested. The determining may be by any means for detecting the presence of, or quantification of expression, including detection and/or measurement of mRNA or detection and/or quantification of expressed protein. Such detection and/or quantification may involve nucleic acid probing and detection of nucleic acids hybridized to a suitable probe or amplification of target nucleic acids using suitable primers. Detection and/or quantification of expressed protein may involve immunological procedures whereby an antibody to the protein is employed in procedures known in the art.
Medicaments and pharmaceutical compositions for use in this invention will comprise one or more peptides having the activity of CNTF and one or more peptides capable of functioning as a CNTF receptor. Preferably, the peptide functioning as the receptor is a soluble peptide capable of binding CNTF. Pharmaceutical compositions and medicaments of this invention may also comprise one or more pharmaceutically acceptable diluents, carriers, excipients or the like including those known in the art for formulation and facilitation of delivery of peptides to target cells or tissues, including CNS tissue.
By "neurodegenerative disorder" is meant a disorder that is characterized by the death or loss of function of neuronal cells, also known as neurons. In some embodiments, a neurodegenerative disorder is characterized by the death or loss of function of neuronal- associated cells, such as astrocytes or other glial cells. The cell death may occur by apoptosis or by necrosis. A neurogenerative disorder includes, without limitation, substance abuse, trauma, stroke, ischemia, Huntington's disease, Alzheimer's disease, Parkinson's disease, prion diseases, such as variant Creutzfeld- Jakob, amyotrophic lateral sclerosis (ALS), olivopontocerebellar atrophy, epilepsy, seizures, and hypoglycemic encephalopathy. A neurodegenerative disorder also includes any disorder or condition that would benefit from neuroprotection.
"Neuroprotection" refers to, in general, protection of neuronal cells (such as neurons) from injury or degeneration. In some embodiments, neuroprotection refers to protection of any nerve tissue, including neurons and neuronal-associated cells (such as astrocytes or other glial cells) from injury or degeneration. In some embodiments, the injury or degeneration may lead to cell death or loss of function. In some embodiments, the injury or degeneration may result from a general process termed "excitotoxicity," which is mediated by the action of glutamate. In some embodiments, the injury or degeneration may be due to physical injury or trauma; a neurodegenerative disorder, including a hereditary genetic disorder; action of a compound, for example, a toxin or a drug, such as a therapeutic or prophylactic; or any other factor that results in nerve tissue injury or degeneration. In some embodiments, the neuroprotection may extend to at least a portion of affected or injured nerve tissue. For example, in stroke, while neurons within the core area of the ischemic insult may be unsalvageable, neurons within the surrounding penumbra have the potential to be rescued, since most neurons within the penumbra do not succumb to the ischemic insult until days following the stroke, and may benefit from the neuroprotective compositions of the invention. In general, the compounds, including test compounds, or combinations of the invention, for example, a CNTF/soluble CNTF receptor combination, exhibit any integer between 10% and 90%, for example, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, or over 100%, neuroprotection when compared to no treatment, or treatment with either CNTF or soluble CNTF receptor alone. "Modulating" or "modulates" means changing, by either increase or decrease. The increase or decrease may be a change of any integer value between 10% and 90%, or of any integer value between 30% and 60%, or may be over 100%, when compared with a control or reference sample or compound.
A "test compound" is any naturally-occurring or artificially-derived chemical compound. Test compounds may include, without limitation, peptides, polypeptides, synthesised organic molecules, naturally occurring organic molecules, and nucleic acid molecules. A test compound can "compete" with a known compound or combination such as a CNTF/soluble CNTF receptor combination by, for example, interfering with upregulation of connexin protein or mRNA expression levels, e.g., Cx43, Cxs 26, Cxs 30, Cxs 31.1, or Cxs 40 levels, by interfering with the action of gap junction blockers, or by interfering with any biological response induced by the known combination, such as neuroprotection. Generally, a test compound can exhibit any value between 10% and 200%), or over 500%, modulation when compared to a CNTF/soluble CNTF receptor combination, or other reference compound. For example, a test compound may exhibit at least any positive or negative integer from 10% to 200% modulation, or at least any positive or negative integer from 30% to 150% modulation, or at least any positive or negative integer from 60% to 100% modulation, or any positive or negative integer over 100% modulation. A compound that is a negative modulator will in general decrease modulation relative to a known compound, while a compound that is a positive modulator will in general increase modulation relative to a known compound.
By "contacting" is meant to submit an animal, cell, lysate, extract, molecule derived from a cell, or synthetic molecule to a test compound.
By "determining" is meant analysing the effect of a test compound on the test system. The means for analysing may include, without limitation, polymerase chain reaction, intercellular coupling assays, immunological assays (e.g, immunoprecipitation, immunofluorescence, ELISA, Western blotting), ultrastructural analysis, histological analysis, kinase assays, cell death assays, Northern or Southern blotting, gene array assays, animal models, or any other methods described herein or known to those skilled in the art.
The terms "nucleic acid" or "nucleic acid molecule" encompass both RNA (plus and minus strands) and DNA, including cDNA, genomic DNA, and synthetic (e.g., chemically synthesized) DNA. The nucleic acid may be double-stranded or single-stranded. Where single-stranded, the nucleic acid may be the sense strand or the antisense strand. A nucleic acid molecule may be any chain of two or more covalently bonded nucleotides, including naturally occurring or non-naturally occurring nucleotides, or nucleotide analogs or derivatives. By "RNA" is meant a sequence of two or more covalently bonded, naturally occurring or modified ribonucleotides. One example of a modified RNA included within this term is phosphorothioate RNA. By "DNA" is meant a sequence of two or more covalently bonded, naturally occurring or modified deoxyribonucleotides. By "cDNA" is meant complementary or copy DNA produced from an RNA template by the action of RNA-dependent DNA polymerase (reverse transcriptase). Thus a "cDNA clone" means a duplex DNA sequence complementary to an RNA molecule of interest, carried in a cloning vector. By "complementary" is meant that two nucleic acids, e.g., DNA or RNA, contain a sufficient number of nucleotides which are capable of forming Watson-Crick base pairs to produce a region of double-strandedness between the two nucleic acids. Thus, adenine in one strand of DNA or RNA pairs with thymine in an opposing complementary DNA strand or with uracil in an opposing complementary RNA strand. It will be understood that each nucleotide in a nucleic acid molecule need not form a matched Watson-Crick base pair with a nucleotide in an opposing complementary strand to form a duplex. A nucleic acid molecule is "complementary" to another nucleic acid molecule if it hybridizes, under conditions of high stringency, with the second nucleic acid molecule. The level of nucleic acid molecule expression may be measured by any technique known in the art, for example, by quantification of hybridization signals. A change in the level of nucleic acid molecule expression may a change of any value between 10% and 200%, for example a change of 10%, 30%, 50%, 100%, or 150%, or over 500%, when compared to a reference system or compound. A "microarray" generally refers to a high density nucleic acid molecule array, which may for example be used to monitor the presence or level of expression of a large number of genes or for detecting sequence variations, mutations and polymorphisms. Microarrays generally require a solid support (for example, nylon, glass, ceramic, plastic, etc.) to which the nucleic acid molecules are attached in a specified 2-dimensional arrangement, such that the pattern of hybridization to a probe is easily determinable.
DETAILED DESCRIPTION
The invention provides, in part, methods for neuroprotection using a "Complex" or a combination of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that as a receptor for CNTF, e.g., CNTFR-alpha, or a biologically-active fragment or variant thereof. The Complex can increase cellular communication among CNS cells, and therefore increases the resilience of CNS cells to insults or injury. Neuroprotection using the Complex or combination of the invention is more effective than that achieved in the absence of treatment (i.e., without applying exogenous agents) or by treatment with CNTF alone, or with soluble CNTF receptor alone.
Polypeptides And Test Compounds Compounds for use in this invention include, without limitation, a peptide having the activity of CNTF; a peptide having the activity or function of a CNTF receptor, e.g., CNTFR- alpha; and combinations thereof. Narious peptides and sequences of peptides having the activity of CΝTF are known in the art and additional variants may be prepared by persons of skill in the art or as described herein. Examples of prior literature describing CΝTF and homologous peptides include WO 91/04316 and United States Patents 4,997,929, 5,011,914 and 5,426,177. Narious CΝTF peptides are described in Accession numbers: ΝP_000605 (human); NP_740756 (mouse); AAH27539 (mouse); NP_443733 (mouse); NP_037298 (rat); UNRBCF (rabbit); P26441 (human); AAM92576 (rhesus monkey); O02732 (pig); P51642 (mouse); Q02011 (chicken); P20294 (rat); or P14188 (rabbit). Antibodies to CNTF are commercially available, for example from Chemicon International, Temecula, California, U.S.A. Narious peptides and sequences of peptides having the activity of a CΝTF receptor, including its soluble portion, are known in the art and additional variants may be prepared by persons of skill in the art or as described herein. Examples of prior literature describing CΝTF receptors, including their soluble portions and homologous peptides include Davis, et al. (1991); and United States Patents 5,426,177 and 5,849,897. Narious CΝTF receptor peptides are described in Accession numbers: ΝP_001833 (human); NP_671693 (human); P51641 (chicken); P26992 (human); Q08406 (rat); 158141 (rat); UHHUCN (human); UNRTCF (rat); or UNHUCF (human). A soluble peptide having the activity of a CNTF receptor may be obtained commercially, for example, from R&D Systems, Minneapolis, MN, USA. A soluble peptide having the activity of a CNTF receptor may also be obtained by, for example, cleaving the membrane linkage of a CNTF receptor using a phospholipase, or by recombinant techniques whereby a peptide having the activity of a CNTF receptor is mutated so that it is no longer capable of associating with membranes. In general, a peptide having the activity of a CNTF receptor is capable of binding CNTF. In embodiments, a peptide having the activity of a CNTF receptor is also capable of binding gpl30 and/or the LIF receptor. Antibodies to the receptor are known, including those described in United States Patent 5,892,003. The CNTF and/or CNTF receptor may be human, or may be derived from any other animal, such as mouse, rat, rabbit, chicken, pig, or monkey. In some embodiments, the invention excludes polypeptide compounds composed of a fusion between CNTF and a CNTF receptor, for example, a full length CNTF receptor. In some embodiments, the invention includes polypeptides including CNTF fused with a soluble CNTF receptor. A "protein," "peptide" or "polypeptide" is any chain of two or more amino acids, including naturally occurring or non-naturally occurring amino acids or amino acid analogues, regardless of post-translational modification (e.g., glycosylation or phosphorylation). An "amino acid sequence", "polypeptide", "peptide" or "protein" of the invention may include peptides or proteins that have abnormal linkages, cross links and end caps, non-peptidyl bonds or alternative modifying groups. Such modified peptides are also within the scope of the invention. The term "modifying group" is intended to include structures that are directly attached to the peptidic structure (e.g., by covalent coupling), as well as those that are indirectly attached to the peptidic structure (e.g., by a stable non-covalent association or by covalent coupling to additional amino acid residues, or mimetics, analogues or derivatives thereof, which may flank the core peptidic structure). For example, the modifying group can be coupled to the amino-terminus or carboxy-terminus of a peptidic structure, or to a peptidic or peptidomimetic region flanking the core domain. Alternatively, the modifying group can be coupled to a side chain of at least one amino acid residue of a peptidic structure, or to a peptidic or peptido- mimetic region flanking the core domain (e.g., through the epsilon amino group of a lysyl residue(s), through the carboxyl group of an aspartic acid residue(s) or a glutamic acid residue(s), through a hydroxy group of a tyrosyl residue(s), a serine residue(s) or a threonine residue(s) or other suitable reactive group on an amino acid side chain). Modifying groups covalently coupled to the peptidic structure can be attached by means and using methods well known in the art for linking chemical structures, including, for example, amide, alkylamino, carbamate or urea bonds. A "biologically-active fragment" of CNTF or a peptide having the activity or function of a CNTF receptor, e.g., CNTFR-alpha, includes an amino acid sequence found in a naturally-occurring CNTF or a soluble CNTF receptor peptide that is capable of neuroprotection as described herein or known to those of ordinary skill in the art. A "variant" of CNTF or a peptide having the activity or function of a CNTF receptor, e.g., CNTFR-alpha, includes a modification, for example, by deletion, addition, or substitution, of an amino acid sequence found in a naturally-occurring CNTF or a soluble CNTF receptor peptide that is capable of neuroprotection as described herein or known to those of ordinary skill in the art. Compounds can be prepared by, for example, replacing, deleting, or inserting an amino acid residue at any position of CNTF or a peptide having the activity or function of a CNTF receptor, e.g., CNTFR-alpha, with other conservative amino acid residues, i.e., residues having similar physical, biological, or chemical properties, and screening for the ability of the compound to modulate neuroprotection.
It is well known in the art that some modifications and changes can be made in the structure of a polypeptide without substantially altering the biological function of that peptide, to obtain a biologically equivalent polypeptide. In one aspect of the invention, polypeptides of the present invention also extend to biologically equivalent peptides that differ from a portion of the sequence of the polypeptides of the present invention by conservative amino acid substitutions.
As used herein, the term "conserved amino acid substitutions" refers to the substitution of one amino acid for another at a given location in the peptide, where the substitution can be made without substantial loss of the relevant function. In making such changes, substitutions of like amino acid residues can be made on the basis of relative similarity of side-chain substituents, for example, their size, charge, hydrophobicity, hydrophilicity, and the like, and such substitutions may be assayed for their effect on the function of the peptide by routine testing.
As used herein, the term "amino acids" means those L-amino acids commonly found in naturally occurring proteins, D-amino acids and such amino acids when they have been modified. Accordingly, amino acids of the invention may include, for example: 2-Aminoadipic acid; 3-Aminoadipic acid; beta-Alanine; beta-Aminopropionic acid; 2-Aminobutyric acid; 4- Aminobutyric acid; piperidinic acid; 6-Aminocaproic acid; 2-Aminoheptanoic acid; 2- Aminoisobutyric acid; 3-Aminoisobutyric acid; 2-Aminopimelic acid; 2,4 Diaminobutyric acid; Desmosine; 2,2'-Diaminopimelic acid; 2,3-Diaminopropionic acid; N-Ethylglycine; N- Ethylasparagine; Hydroxylysine; allo-Hydroxylysine; 3-Hydroxyproline; 4-Hydroxyproline; Isodesmosine; allo-Isoleucine; N-Methylglycine; sarcosine; N-Methylisoleucine; 6-N- methyllysine; N-Methylvaline; Norvaline; Norleucine; and Ornithine.
In some embodiments, conserved amino acid substitutions may be made where an amino acid residue is substituted for another having a similar hydrophilicity value (e.g., within a value of plus or minus 2.0, or plus or minus 1.5, or plus or minus 1.0, or plus or minus 0.5), where the following may be an amino acid having a hydropathic index of about -1.6 such as Tyr (-1.3) or Pro (-1.6) are assigned to amino acid residues (as detailed in United States Patent No. 4,554,101, incorporated herein by reference): Arg (+3.0); Lys (+3.0); Asp (+3.0); Glu (+3.0); Ser (+0.3); Asn (+0.2); Gin (+0.2); Gly (0); Pro (-0.5); Thr (-0.4); Ala (-0.5); His (-0.5); Cys (-1.0); Met (-1.3); Nal (-1.5); Leu (-1.8); lie (-1.8); Tyr (-2.3); Phe (-2.5); and Trp (-3.4). In alternative embodiments, conservative amino acid substitutions may be made where an amino acid residue is substituted for another having a similar hydropathic index (e.g., within a value of plus or minus 2.0, or plus or minus 1.5, or plus or minus 1.0, or plus or minus 0.5). In such embodiments, each amino acid residue may be assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics, as follows: He (+4.5); Nal (+4.2); Leu (+3.8); Phe (+2.8); Cys (+2.5); Met (+1.9); Ala (+1.8); Gly (-0.4); Thr (-0.7); Ser (-0.8); Trp (-0.9); Tyr (-1.3); Pro (-1.6); His (-3.2); Glu (-3.5); Gin (-3.5); Asp (-3.5); Asn (-3.5); Lys (-3.9); and Arg (-4.5).
In alternative embodiments, conservative amino acid substitutions may be made using publicly available families of similarity matrices (Altschul, S.F. 1991. "Amino acid substitution matrices from an information theoretic perspective." Journal of Molecular Biology, 219: 555- 665; Dayhoff, M.O., Schwartz, R.M., Orcutt, B.C. 1978. "A model of evolutionary change in proteins." In "Atlas of Protein Sequence and Structure" 5(3) M.O.
Dayhoff (ed.), 345 - 352, National Biomedical Research Foundation, Washington; States, D.J., Gish, W., Altschul, S.F. 1991. "Improved Sensitivity of Nucleic Acid Database Search Using Application-Specific Scoring Matrices" Methods: A companion to Methods in Enzymology 3(1): 66 - 77; Steven Henikoff and Jorja G. Henikoff. 1992 "Amino acid substitution matrices from protein blocks." Proc. Natl. Acad. Sci. USA. 89(biochemistry): 10915 - 10919; M.S. Johnson and J.P. Overington. 1993. "A Structural Basis of Sequence Comparisons: An evaluation of scoring methodologies." Journal of Molecular Biology. 233: 716 - 738. Steven Henikoff and Jorja G. Henikoff. 1993. "Performance Evaluation of Amino Acid Substitution Matrices." Proteins: Structure, Function, and Genetics. 17: 49- 61; Karlin, S. and Altschul, S.F. 1990. "Methods for assessing the statistical significance of molecular sequence features by using general scoring schemes" Proc. Natl. Acad. Sci. USA. 87: 2264 - 2268.) The PAM matrix is based upon counts derived from an evolutionary model, while the Blosum matrix uses counts derived from highly conserved blocks within an alignment. A similarity score of above zero in either of the PAM or Blosum matrices may be used to make conservative amino acid substitutions.
In alternative embodiments, conservative amino acid substitutions may be made where an amino acid residue is substituted for another in the same class, where the amino acids are divided into non-polar, acidic, basic and neutral classes, as follows: non-polar: Ala, Nal, Leu, He, Phe, Trp, Pro, Met; acidic: Asp, Glu; basic: Lys, Arg, His; neutral: Gly, Ser, Thr, Cys, Asn, Gin, Tyr.
Conservative amino acid changes can include the substitution of an L-amino acid by the corresponding D-amino acid, by a conservative D-amino acid, or by a naturally-occurring, non- genetically encoded form of amino acid, as well as a conservative substitution of an L-amino acid. Naturally-occurring non-genetically encoded amino acids include beta-alanine, 3-amino- propionic acid, 2,3-diamino propionic acid, alpha-aminoisobutyric acid, 4-amino-butyric acid, N-methylglycine (sarcosine), hydroxyproline, ornithine, citrulline, t-butylalanine, t- butylglycine, N-methylisoleucine, phenylglycine, cyclohexylalanine, norleucine, norvaline, 2- napthylalanine, pyridylalanine, 3-benzothienyl alanine, 4-chlorophenylalanine, 2- fluorophenylalanine, 3-fluorophenylalanine, 4-fluorophenylalanine, penicillamine, 1,2,3,4- tetrahydro-isoquinoline-3-carboxylix acid, beta-2-thienylalanine, methionine sulfoxide, homoarginine, N-acetyl lysine, 2-amino butyric acid, 2-amino butyric acid, 2,4,-diamino butyric acid, p-aminophenylalanine, N-methylvaline, homocysteine, homoserine, cysteic acid, epsilon-amino hexanoic acid, delta-amino valeric acid, or 2,3-diaminobutyric acid.
In alternative embodiments, conservative amino acid changes include changes based on considerations of hydrophilicity or hydrophobicity, size or volume, or charge. Amino acids can be generally characterized as hydrophobic or hydrophilic, depending primarily on the properties of the amino acid side chain. A hydrophobic amino acid exhibits a hydrophobicity of greater than zero, and a hydrophilic amino acid exhibits a hydrophilicity of less than zero, based on the normalized consensus hydrophobicity scale of Eisenberg et al(J. Mol. Bio. 179:125-142, 184). Genetically encoded hydrophobic amino acids include Gly, Ala, Phe, Nal, Leu, He, Pro, Met and Trp, and genetically encoded hydrophilic amino acids include Thr, His, Glu, Gin, Asp, Arg, Ser, and Lys. Νon-genetically encoded hydrophobic amino acids include t-butylalanine, while non-genetically encoded hydrophilic amino acids include citrulline and homocysteine.
Hydrophobic or hydrophilic amino acids can be further subdivided based on the characteristics of their side chains. For example, an aromatic amino acid is a hydrophobic amino acid with a side chain containing at least one aromatic or heteroaromatic ring, which may contain one or more substituents such as -OH, -SH, -CΝ, -F, -CI, -Br, -I, -ΝO2, -NO, - NH2, -NHR, -NRR, -C(O)R, -C(O)OH, -C(O)OR, -C(O)NH2, -C(O)NHR, -C(O)NRR, etc., where R is independently (Cι-C6) alkyl, substituted (Cι-C6) alkyl, (Cι-C6) alkenyl, substituted (Cι-C6) alkenyl, (Cι-C6) alkynyl, substituted (Ci-C6) alkynyl, (C5-C2o) aryl, substituted (C5- C2o) aryl, (C6-C26) alkaryl, substituted (C6-C26) alkaryl, 5-20 membered heteroaryl, substituted 5-20 membered heteroaryl, 6-26 membered alkheteroaryl or substituted 6-26 membered alkheteroaryl. Genetically encoded aromatic amino acids include Phe, Tyr, and Trp, while non-genetically encoded aromatic amino acids include phenylglycine, 2-napthylalanine, beta-2- thienylalanine, 1, 2,3 ,4-tetrahydro-isoquinoline-3 -carboxylic acid, 4-chlorophenylalanine, 2- fluorophenylalanine3 -fluorophenylalanine, and 4-fluorophenylalanine.
An apolar amino acid is a hydrophobic amino acid with a side chain that is uncharged at physiological pH and which has bonds in which a pair of electrons shared in common by two atoms is generally held equally by each of the two atoms (i.e., the side chain is not polar). Genetically encoded apolar amino acids include Gly, Leu, Nal, He, Ala, and Met, while non- genetically encoded apolar amino acids include cyclohexylalanine. Apolar amino acids can be further subdivided to include aliphatic amino acids, which is a hydrophobic amino acid having an aliphatic hydrocarbon side chain. Genetically encoded aliphatic amino acids include Ala, Leu, Nal, and He, while non-genetically encoded aliphatic amino acids include norleucine. A polar amino acid is a hydrophilic amino acid with a side chain that is uncharged at physiological pH, but which has one bond in which the pair of electrons shared in common by two atoms is held more closely by one of the atoms. Genetically encoded polar amino acids include Ser, Thr, Asn, and Gin, while non-genetically encoded polar amino acids include citrulline, Ν-acetyl lysine, and methionine sulfoxide.
An acidic amino acid is a hydrophilic amino acid with a side chain pKa value of less than 7. Acidic amino acids typically have negatively charged side chains at physiological pH due to loss of a hydrogen ion. Genetically encoded acidic amino acids include Asp and Glu. A basic amino acid is a hydrophilic amino acid with a side chain pKa value of greater than 7. Basic amino acids typically have positively charged side chains at physiological pH due to association with hydronium ion. Genetically encoded basic amino acids include Arg, Lys, and His, while non-genetically encoded basic amino acids include the non-cyclic amino acids ornithine, 2,3,-diaminopropionic acid, 2,4-diaminobutyric acid, and homoarginine.
It will be appreciated by one skilled in the art that the above classifications are not absolute and that an amino acid may be classified in more than one category. In addition, amino acids can be classified based on known behaviour and or characteristic chemical, physical, or biological properties based on specified assays or as compared with previously identified amino acids. Amino acids can also include bifunctional moieties having amino acidlike side chains.
Conservative changes can also include the substitution of a chemically derivatised moiety for a non-derivatised residue, by for example, reaction of a functional side group of an amino acid. Thus, these substitutions can include compounds whose free amino groups have been derivatised to amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t- butyloxycarbonyl groups, chloroacetyl groups or formyl groups. Similarly, free carboxyl groups can be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides, and side chains can be derivatized to form O-acyl or O-alkyl derivatives for free hydroxyl groups or N-im-benzylhistidine for the imidazole nitrogen of histidine. Peptide analogues also include amino acids that have been chemically altered, for example, by methylation, by amidation of the C-terminal amino acid by an alkylamine such as ethylamine, ethanolamine, or ethylene diamine, or acylation or methylation of an amino acid side chain (such as acylation of the epsilon amino group of lysine). Peptide analogues can also include replacement of the amide linkage in the peptide with a substituted amide (for example, groups of the formula -C(O)-NR, where R is (Cι-C6) alkyl, (Cι-C6) alkenyl, (Cι-C6) alkynyl, substituted (Ci-Ce) alkyl, substituted (Cι-C6) alkenyl, or substituted (Cι~C6) alkynyl) or isostere of an amide linkage (for example, -CH2NH-, -CH2S, -CH2CH2-, -CH=CH- (cis and trans), - C(O)CH2-, -CH(OH)CH2-, or-CH2SO-). The compound can be covalently linked, for example, by polymerisation or conjugation, to form homopolymers or heteropolymers. Spacers and linkers, typically composed of small neutral molecules, such as amino acids that are uncharged under physiological conditions, can be used. Linkages can be achieved in a number of ways. For example, cysteine residues can be added at the peptide termini, and multiple peptides can be covalently bonded by controlled oxidation. Alternatively, heterobifunctional agents, such as disulfide/amide forming agents or thioether/amide forming agents can be used. The compound can also be linked to a another compound that can modulate a neuroprotective response. The compound can also be constrained, for example, by having cyclic portions.
Peptides or peptide analogues can be synthesised by standard chemical techniques, for example, by automated synthesis using solution or solid phase synthesis methodology.
Automated peptide synthesisers are commercially available and use techniques well known in the art. Peptides and peptide analogues can also be prepared using recombinant DNA technology using standard methods such as those described in, for example, Sambrook, et al. (Molecular Cloning: A Laboratory Manual. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989) or Ausubel et al. (Current Protocols in Molecular Biology, John Wiley & Sons, 1994). In general, candidate compounds are identified from large libraries of both natural products or synthetic (or semi-synthetic) extracts or chemical libraries according to methods known in the art. Those skilled in the field of drug discovery and development will understand that the precise source of test extracts or compounds is not critical to the method(s) of the invention. Accordingly, virtually any number of chemical extracts or compounds can be screened using the exemplary methods described herein. Examples of such extracts or compounds include, but are not limited to, plant-, fungal-, prokaryotic- or animal-based extracts, fermentation broths, and synthetic compounds, as well as modification of existing compounds. Numerous methods are also available for generating random or directed synthesis (e.g., semi-synthesis or total synthesis) of any number of chemical compounds, including, but not limited to, saccharide-, lipid-, peptide-, and nucleic acid-based compounds. Synthetic compound libraries are commercially available. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant, and animal extracts are commercially available from a number of sources, including Biotics (Sussex, UK), Xenova (Slough, UK), Harbor Branch Oceanographic Institute (Ft. Pierce, FL, USA), and PharmaMar, MA, USA. In addition, natural and synthetically produced libraries of, for example, candidate polypeptides, are produced, if desired, according to methods known in the art, e.g., by standard extraction and fractionation methods. Furthermore, if desired, any library or compound is readily modified using standard chemical, physical, or biochemical methods.
When a crude extract is found to modulate neuroprotection, further fractionation of the positive lead extract is necessary to isolate chemical constituents responsible for the observed effect. Thus, the goal of the extraction, fractionation, and purification process is the careful characterization and identification of a chemical entity within the crude extract having neuroprotective activities. The same assays described herein for the detection of activities in mixtures of compounds can be used to purify the active component and to test derivatives thereof. Methods of fractionation and purification of such heterogeneous extracts are known in the art. If desired, compounds shown to be useful agents for treatment are chemically modified according to methods known in the art. Compounds identified as being of therapeutic, prophylactic, diagnostic, or other value may be subsequently analyzed using a murine stroke model, or any other animal model suitable for neuroprotection studies, such as animal models of neurodegenerative disease (see, for example, Jankowsky JL et al, 2002, Transgenic mouse models of neurodegenerative disease: opportunities for therapeutic development, Curr Neurol Neurosci Rep. Sep;2(5):457-64; Hodgson et al. (1999) Neuron 23, 181-192).
Pharmaceutical & Veterinary Compositions, Dosages, And Administration The combinations or compounds of the invention can be provided alone or in combination with other compounds (for example, nucleic acid molecules, small molecules, peptides, or peptide analogues), in the presence of a liposome, an adjuvant, or any pharmaceutically acceptable carrier, in a form suitable for administration to mammals, for example, humans. If desired, treatment with a compound according to the invention may be combined with more traditional and existing therapies for providing neuroprotection, or for treating or prevention a neurodegenerative disorder. In some embodiments, the CNTF peptide, or biologically-active fragment or variant thereof, may be administered in the same formulation as the soluble peptide having the activity or function of a CNTF receptor, e.g., CNTFR-alpha, or biologically-active fragment or variant thereof, or may be administered as separate formulations. If administered in the same formulation, in some embodiments, the CNTF peptide, or biologically-active fragment or variant thereof, and the soluble peptide having the activity or function of a CNTF receptor, e.g., CNTFR-alpha, or biologically-active fragment or variant thereof, may be administered as a complex, for example, as a covalently bound complex, or a non-covalently bound complex. In general, a "complex" refers to a combination in which the relevant ingredients are capable interacting, such as binding of CNTF to a soluble peptide or compound having the activity or function of a CNTF receptor.
Conventional pharmaceutical practice may be employed to provide suitable formulations or compositions to administer the compounds to subjects suffering from or presymptomatic for a neurodegenerative disorder, or in need of neuroprotection. Any appropriate route of administration may be employed, for example, parenteral, intravenous, subcutaneous, intramuscular, intracranial, intraorbital, ophthalmic, intraventricular, intracapsular, intraspinal, intrathecal, intracisternal, intraperitoneal, intranasal, aerosol, or oral administration. Therapeutic formulations maybe in the form of liquid solutions or suspensions; for oral administration, formulations may be in the form of tablets or capsules; and for intranasal formulations, in the form of powders, nasal drops, or aerosols. In embodiments, the compounds of the invention are delivered directly or indirectly to CNS tissue. Methods for formulation and delivery of peptides to CNS tissue are known, including methods for administering CNTF to the brain as described in United States Patent application 10/073,658 published June 13, 2002.
Methods well known in the art for making formulations are found in, for example, "Remington's Pharmaceutical Sciences" (19th edition), ed. A. Gennaro, 1995, Mack Publishing Company, Easton, Pa. Formulations for parenteral administration may, for example, contain excipients, sterile water, or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated napthalenes. Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds. Other potentially useful parenteral delivery systems for modulatory compounds include ethylene- vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel. For therapeutic or prophylactic compositions, the compounds are administered to an individual in an amount sufficient to stop or slow neurodegeneration or to provide neuroprotection, depending on the disorder. An "effective amount" of a compound according to the invention includes a therapeutically effective amount or a prophylactically effective amount. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as neuroprotection, or the inhibition or prevention of neurodegeneration. A therapeutically effective amount of a compound may vary- according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result, such as neuroprotection, or the inhibition or prevention of neurodegeneration. Typically, a prophylactic dose is used in subjects prior to or at an earlier stage of disease, so that a prophylactically effective amount may be less than a therapeutically effective amount. A preferred range for therapeutically or prophylactically effective amounts of a compound may be any integer from 0.1 nM-O.lM, 0.1 nM-0.05M, 0.05 nM-15μM or 0.01 nM-10μM. It is to be noted that dosage values may vary with the severity of the condition to be alleviated. For any particular subject, specific dosage regimens maybe adjusted over time according to the individual need and the professional judgement of the person administering or supervising the administration of the compositions. Dosage ranges set forth herein are exemplary only and do not limit the dosage ranges that may be selected by medical practitioners. The amount of active compound in the composition may vary according to factors such as the disease state, age, sex, and weight of the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
In the case of vaccine formulations, an immunogenically effective amount of a compound of the invention can be provided, alone or in combination with other compounds, with an immunological adjuvant, for example, Freund's incomplete adjuvant, NSA3, dimethyldioctadecylammonium hydroxide, or aluminum hydroxide. The compound may also be linked with a carrier molecule, such as bovine serum albumin or keyhole limpet hemocyanin to enhance immunogenicity.
In general, compounds of the invention should be used without causing substantial toxicity. Toxicity of the compounds of the invention can be determined using standard techniques, for example, by testing in cell cultures or experimental animals and determining the therapeutic index, i.e., the ratio between the LD50 (the dose lethal to 50% of the population) and the LD100 (the dose lethal to 100% of the population). In some circumstances however, such as in severe disease conditions, it may be necessary to administer substantial excesses of the compositions.
The following examples are meant to illustrate embodiments of the invention and should not be construed as limiting the scope of the invention. EXAMPLE A Increasing Connexin 43 Expression in Glioma Cells Cell Culture
C6 glioma cells, obtained from American Type Culture Collection, were grown in DMEM supplemented with 10% (v/v) fetal bovine serum (FBS), 10 μg/ml streptomycin and 10 units/ml penicillin. Twenty-four hours prior to and throughout, all cells were maintained in serum-reduced media (1% FBS). Where noted, cells were exposed to the following agents: vehicle (phosphate buffered saline, PBS), CNTF (20 ng/ml), CNTFRα (200 ng/ml) or Complex (CNTF + CNTFRα).
Total RNA Isolation, cDNA Preparation and PCR
C6 cells were washed twice with PBS and total RNA was extracted using the phenol- chloroform-isoamyl alcohol method outlined by Sambrook et al. (1989). Reverse transcription- polymerase chain reaction (RT-PCR) of the RNA was performed to create cDNA according to the method obtained with supplies by Invitrogen Corp. (Burlington, ON). Briefly, 3 μg of RNA was pre-treated with DNAse and then reverse transcribed in a thermal cycler (Perkin Elmer, Norwalk, CT) using Superscript II and oligo-dT primers for 1 h at 42°C. The cDNA product (1 μl) was mixed with Tris-HCl (pH 8.4; 20 mM), MgCl2 (1.5 mM), dNTP (200 μM), Platinum Taq (2 U), and one set of oligonucleotide primers (200 μM) given in final concentrations in a final volume of 40 μl. Samples were denatured for 5 min at 95 °C and then amplified for 30 cycles of 94°C/45 sec, 58°C/1 min, and 72°C/1 min. Twenty microlitres of each PCR sample was run on a 1.8 % agarose gel in parallel with a 1.5 kb DNA standard (Invitrogen Corp., Burlington, ON).
Primers for CNTF, CNTFRα and glyceraldehyde-3 -phosphate dehydrogenase (GAPDH; used as a positive RNA-loading control) were used for amplification of the cDNA. Primer sequences are shown in Table 1.
False positive reactions were ruled out by omitting Superscript II from the cDNA preparation. Primers that produced negative results were subjected to a positive control by performing the RT-PCR on brain tissue (cortex and cerebellum) isolated from an adult CD-I mouse. Table 1. Primer sequences used for PCR analysis of cDNA
Target Primers SEQ Expected Reference ID Product NO: Size
CNTF Forward 5 '-GGAAGATTCGTTCAGACCTGAC-3' 1 350 bp (Malgrange et α/.,
Reverse 5 '-CCCATCAGCCTCATTTTCAGGG-3' 2 1998)
CNTFRα Forward 5 '-CCACATGCTGCCATTGATCC-3 ' 3 430 bp (Malgrange et α/., Reverse 5 '-CCACATGCTGCCATTGATCC-3 ' 4 1998)
Cx43 Forward 5 '-CCCCACTCTCACCTATGTCTCC-3 ' 5 500 bp (Naus et al.,
Reverse 5 '-ACTTTTGCCGCCTAGCTATCCC-3 ' 6 1997)
Cx30 Forward 5^AATGTGGCCGAGTTGTGTTA-3' 399 bp (Dahl et βt., 1996)
Reverse 5'-CCAAGGCCCAGTTGTCAC-3'
GAPDH Forward 5 '-AATGCATCCTGCACCACCAA-3' 9 515 bp (Yang et al.,
Reverse 5 '-GTAGCCATATTCATTGTCATA-3 ' 10 2001)
Immunocytochemistry
Cells were grown to 80% confluence and subsequently treated with agents every 24 h (with fresh media changes) for three days. Cells were then washed twice with PBS and fixed for 10 minutes with 70% ethanol containing 0.15 M NaCl. Following blockage of non-specific antibody binding using 10% normal goat serum in PBS for 1 h, cells were incubated with rabbit polyclonal anti-Cx43 antibody (1 :400 dilution, Sigma- Aldrich, Oakville, ON) for 1 h, rinsed with PBS and subsequently incubated with Alexa-Fluor-conjugated goat anti-rabbit IgG secondary antibody (1 :500 dilution, Molecular Probes, Eugene, OR). Cells were then washed with PBS, stained with Hoechst 33342 dye (20 ng/ml, Sigma-Aldrich), and mounted with Nectashield medium (Nector Laboratories, Inc. Burlingame, CA). Staining was visualized with a Zeiss Axiophot photomicroscope (Carl Zeiss, Thornwood, ΝY) and captured using Northern Exposure, version 2 (ImageExperts Inc., Mississauga, ON). Protein Isolation and Western Blot Analysis of Cx43
C6 cells were treated with agents every 24 h (with fresh media changes) for three days, rinsed twice with PBS and scraped off the plates in lysis buffer (RIP A buffer supplemented with protease inhibitors (complete, Mini; Roche, Indianapolis, IN )). DNA in the lysate was sheared using a 22 gauge needle. Total cell lysate was collected following microcentrifugation at 10,000xg for 10 minutes and total protein concentration was determined using the BCA Protein Assay Kit (Pierce-BioLynx, Brockville, ON). Samples (50 μg) were run in parallel with molecular weight markers (Bio-Rad Lab., Hercules, CA) on a 10%o sodium dodecyl sulfate- polyacrylamide gel by electrophoresis. Protein bands were transferred to a nitrocellulose membrane at 80 watts for 1 h and subsequently blocked with 5% nonfat dry milk in PBS (with 1%) Tween-20) for 1 h. Following three PBS rinses (10 minutes each), the membrane was incubated in anti-Cx43 antibody (1:400 dilution, Sigma- Aldrich) for 1 h, rinsed again with PBS, and then bathed in secondary antibody tagged with horseradish peroxidase (1 :20,000 dilution, CedarLane Lab. Ltd., Hornby, ON). Following three PBS rinses, the membrane was incubated in Supersignal (Pierce-BioLynx) and exposed to X-ray film to visualize antibody binding. To normalize protein loading the membranes were gently stripped of antibodies and immunoblotted for GAPDH (1: 20,000 dilution, CedarLane Lab. Ltd.).
Pre-Loading Gap junctional coupling of C6 cells was determined by the pre-loading method as described by Goldberg et al. (1995). Briefly, C6 cells were grown to confluence in 12-well plates. Twenty-four h prior to pre-loading, a media change containing the various agents was performed. The agents were present in all solutions throughout the procedures. Donor C6 cells were preloaded with dye solution (5 μM calcein-AM (Molecular Probes) and 10 μM Dil (Sigma- Alderich) in an isotonic (0.3 M) glucose solution) for 20 min in a humidified incubator (37°C, 5% CO2/95% air). Subsequently, donor cells were rinsed twice with glucose solution, trypsinized, suspended in growth media, and seeded onto recipient (unlabeled) cells at 1:500 ratio. After being maintained in the incubator for 3 h, cells were examined with a photomicroscope. Gap junctional communication was assessed by the passage of calcein from donor cells to the underlying recipient cells. Only cells that were coupled to at least one other cell were examined. Growth Curves
C6 cells were seeded into 12-well plates at 2 x 104 cells/well (designated Day -1). Cells received fresh media containing the various agents every 24 h commencing on Day 0. The number of cells in each well were counted on Days 0-4, 6 and 8 using a haemocytometer and trypan blue as a dilutant.
Data Analysis
Results were expressed as means ± standard error of the means of four or more independent procedures. Statistical comparisons were performed using one way analysis of variance with a P value of <0.05 considered significant.
C6 Cells Endogenously Express CNTF
To determine if C6 cells have endogenous expression of mRNA for either CNTF or CNTFRα, RT-PCR was performed on total RNA. Compared to GAPDH levels, a low amount of CNTF mRNA was present in C6 cells while CNTFRα mRNA could not be detected. Specificity of the CNTFRα primers was confirmed on adult mouse brain samples.
Complex Induces Upregulation of Cx43 in C6 Cells
Confluent cultures were exposed to vehicle, CNTF, CNTFRα or Complex every 24 h for 3 days to determine whether Cx43 is influenced by the CNTF pathway in C6 cells.
Compared to vehicle, CNTF and CNTFRα alone did not alter Cx43 protein expression. In contrast, immunoblot analysis indicated that Cx43 protein levels increased in Complex-treated cells (compared to vehicle). This effect of Complex on Cx43 protein levels predominately occurred by causing an increase in the non-phosphorylated form of the connexin. When protein loading was normalized against GAPDH levels, Complex increased Cx43 levels to 157 % that of vehicle, CNTF and CNTFRα (n = 4; P<0.01). No significant differences in Cx43 protein expression was detected between vehicle, CNTF or CNTFRα treatments.
C6 cell cultures contain a mixed population of cells in regards to protein expression (Roser et al, 1991). When examining Cx43 expression by immunocytochemistry, a heterogenous population of Cx43 -expressing cells was identified; some cells showed low Cx43 staining (-70% of the population) and others with very low staining (the remaining 30% of the population). While no detectable changes in the ratios of this heterogenous population were induced by CNTF, CNTFRα or Complex, immunocytochemical analysis revealed that Complex induced an increase in Cx43 compared to vehicle, CNTF and CNTFRα alone. This increase in Cx43 was detected in both the cytoplasm and at the cell membrane.
Complex Increases Coupling in C6 Cells Dye coupling of C6 cells was examined to determine if Complex-induced upregulation of Cx43 also increased gap junctional communication. No apparent change in the initial coupling of the pre-loaded cells was detected between vehicle, CNTF, CNTFRα or Complex treatments; ~ 75% of donor cells were coupled to at least one recipient cell for all treatments in the allowed time. However, when the number of recipient cells coupled to a single donor cell was examined, a significant difference between the treatments was observed. While neither CNTF nor CNTFRα alone affected coupling compared to vehicle, Complex significantly increased intercellular dye coupling based on the passage of the gap junction permeable dye calcein. When quantified, Complex significantly increased the number of recipient cells coupled to one donor cell from 4-7 cells (vehicle, CNTF, CNTFRα) to 41 cells (n=4; PO.001).
Complex Reduces C6 Cell Proliferation
When C6 cells were grown in serum-reduced media in the presence of the agents, Complex had significantly retarded the growth rate by Day 4 compared to vehicle, CNTF, and CNTFRα alone. The reduction in proliferation induced by Complex became more evident by Day 6, when cultures treated with vehicle, CNTF or CNTFRα alone had reached confluence whereas Complex-treated cultures had not. The significant decrease in C6 cell growth rate induced by Complex was not attributed to any toxic effects of the agent, since no cell death was detected by trypan blue exclusion.
Summary
The C6 glioma cell line expresses the transcript for CNTF but not CNTFRα. When CNTF is administered in the presence of the soluble CNTFRα, an increase in Cx43 and gap junctional communication is detected in C6 cells, accompanied by a significant decrease in C6 cell growth rate. Positive identification of CNTF mRNA in C6 glioma cells shows that this glioma is astrocytic in origin. Furthermore, examination for CNTF provides an alternative method in determining whether a neoplastic cell has an astrocytic lineage. This identification technique is highly useful, since the vast majority of astrocytomas, including both cell lines and in vivo biopsies, lack glial fibrillary acidic protein expression, a marker commonly used to identify and distinguish astrocytes from other CNS cell types (Deck et al, 1978).
The lack of CNTFRα mRNA in C6 glioma cells shows that C6 glioma cells are not immortalized reactive astrocytes. Although C6 cells lack CNTFRα, they respond to CNTF if co-administered with CNTFRα. The increase of Cx43 in C6 cells induced by Complex shows that the CNTF signaling pathway can overcome the Cx43 expression deficiency in these cells.
EXAMPLE B Increased Connexin 43 Expression and Intracellular Coupling in Astrocytes
Astrocyte Cultures
Primary cultures of murine cortical astrocytes were prepared in a similar manner to that described by Fedoroff and Richardson (1997). Briefly, brains were removed from 1 -day-old CD-I mouse pups and subsequently freed of meninges. Cortices were isolated, placed into growth media (DME media supplemented with 10% FBS, 10 units/ml penicillin, and 10 μg/ml streptomycin; Invitrogen Corp.), and mechanically dissociated using a serological pipette. The cell suspension was then passed through a 70 μm cell strainer (Falcon, NWR International) and subsequently diluted with growth media at a ratio of 5 ml/cortex. Cells were plated onto 60 mm dishes (3 ml cell suspension) or 100 mm dishes (10 ml cell suspension) and maintained in a humidified incubator at 37°C in 95% air/5% CO2. Media was changed every 3 days thereafter in addition to shaking the cultures. After 6 weeks, cultures were maintained in Media I (54 ml Νeurobasal Medium (Invitrogen Corp.), 36 ml DMEM/F12 (Invitrogen Corp.), D-glucose (0.6%), insulin (10 μg/ml), transferrin (20 μg/ml), putrescine-HCl (62 μM), progesterone (20 nM), sodium selenite (30 nM), 10 units/ml penicillin, and 10 μg/ml streptomycin) for 1 week. All procedures were performed in Media I.
Exposure to CΝTF and CΝTFRα
Astrocytes were treated with either vehicle (PBS), CΝTF (20 ng/ml; R&D Systems), soluble CΝTFRα (200 ng/ml; R&D Systems), or Complex (20 ng/ml CΝTF + 200 ng/ml CΝTFRα). For expression of Cx43 protein, cells were treated with agents in fresh Media I every 24 hours for a total of 3 days. For examination of mRΝA levels, cells were treated with agents for 24 hours. For assessment of STAT3, ERK1 (p44 MAPK) or ERK2 (p42 MAPK) phosphorylation, cells received media change and 24 hours later, agents were added for 5, 15 and 60 minutes. The STAT3 and MAPK/ERK pathways were inhibited by administering the pharmacological agents AG490 (50 μM; Calbiochem)(Hodge et al, 2002) and U0126 (10 μM; Promega Corp.)(Favata et al, 1998), respectively, 45 minutes prior to adding vehicle or Complex.
Protein Isolation and Immunoblot Analysis
Astrocyte monolayers were rinsed twice with PBS and harvested in radioimmuno- precipitation assay buffer (1% Nonidet P-40, 0.5%ι sodium deoxycholate and 0.1% SDS in PBS) supplemented with protease inhibitors (mini,Complete; Roche) using a rubber policeman. The lysate was sheared using a 22 gauge needle and centrifuged at 10,000 xg for 10 minutes at 4°C. For Cx43 protein analyses, some samples were treated with alkaline phosphatase (calf- intestinal; Roche) to detect total Cx43 protein. Protein concentration of total cell lysate was determined using the BCA Protein Assay Kit (Pierce). Protein samples (50 μg each) and molecular weight markers (Bio-Rad Lab.) were subjected to 10% SDS-polyacrylamide gel electrophoresis and subsequently electro-transferred onto a nitrocellulose membrane for 1 hour at 80 watts. The membrane was immunoblotted using appropriate primary antibodies (Cx43, Sigma; phospho-STAT3 and phospho-p44/42 MAPK, Cell Signaling Tech.) and subsequently incubated in secondary antibodies tagged with horseradish peroxidase (CedarLane Lab. Ltd.). The blots were then incubated in Supersignal (Pierce) and exposed to Kodak X-Omat x-ray film to visualize antibody binding. To normalize protein loading, membranes were stripped of antibodies using 2-βmercaptoethanol (10 mM), SDS (2%), and Tris (62.5 mM, pH 6.7) for 30 min at 65°C and probed for glyceraldehyde-3 -phosphate dehydrogenase (GAPDH; CedarLane Lab. Ltd.), STAT3 (Cell Signaling Tech.), or ERK1 and ERK2 (Santa Cruz Biotechnology, Inc.).
Immunocytochemistry
Astrocytes were rinsed twice with PBS and subsequently fixed with 4% formaldehyde for 10 minutes. Cells were then washed with PBS and blocked for nonspecific binding using 10% normal goat serum in PBS. Subsequently, cells were incubated with primary antibody according to company recommendations (Cx43 polyclonal antibody, Sigma; phospho-STAT3, Cell Signaling). Following three washes with PBS, cells were incubated in Alexa-conjugated secondary IgG (Molecular Probes) for 1 hour and then mounted with Nectashield mounting medium (Nector). Immunostaining was viewed using a Zeiss Axioskop microscope and images were captured using Northern Exposure, Nersion2 (ImageExperts Inc.).
Scrape-Loading/Dye Transfer Assay Intercellular coupling between astrocytes was measured using a modified version of the scrape-loading technique described by el-Fouly et al. (1987). Briefly, media was aspirated from the cultures, cells were bathed in 50 μL of dye solution (0.1 % carboxyfluorescein and 0.1 % dextran tetramethylrhodamine (Molecular Probes) in PBS) and, subsequently, scraped using a surgical blade. After 90 sec, cells were washed several times with PBS containing carbenoxolone (100 μM; Sigma), a gap junction blocker, to halt further progression of carboxyfluorescein through gap junctions and were immediately examined using a Zeiss Axioskop microscope.
RΝA Isolation Cytoplasmic RΝA was isolated from astrocytes using the phenol-chloroform-isoamyl alcohol method outlined by Sambrook et al. (1989). Briefly, cells were washed with PBS and, subsequently, lysed using Lysis Solution (0.2 M ΝaCl, 20 mM MgCl2, and 20 mM Tris pH 8.8). Following centrifugation at 16,000 xg for 30 sec, the supernatant was added to Phenol Solution (0.75 g phenol, 3.4 mM 8-hydroxyquinoline, 4.2 M guanidine thiocyanate, 26.4 mM sodium citrate, 0.5 % sarcosyl, 0.4 % β-mercaptoethanol, and 100 mM sodium acetate) followed by chloroform (10%). The mixture was then centrifuged at 16,000 xg for 15 min and the aqueous layer was added to an equal volume isopropanol. The RΝA was pelleted from the mixture, washed with 90% ethanol and quantified by spectrophotometry at 260 nm.
RT-PCR and Semiquantitative RT-PCR
RT-PCR was performed on the RΝA as for Example A. 3 μg of RΝA was pretreated with DΝAse and subsequently reverse transcribed in a thermal cycler. For semiquantitative RT-PCR, 25 cycles were used to amplify the cDΝA to avoid saturation. Primer sets used for amplification are provided in Table I. The amplified cDΝA was run on a 1.8% agarose gel containing ethidium bromide
(12%) in parallel with a 1 kb DΝA standard (Invitrogen Corp.). To rule out false positives, parallel procedures were performed in the absence of the reverse transcriptase. Functionality of all primers was confirmed by performing RT-PCR on RΝA isolated from brain cortex of adult CD-I mice. Images of amplified products were visualized and captured using Kodak ds ID digital software.
Northern Blot Analysis Cells were exposed to either vehicle, CNTF, CNTFRα, or Complex in fresh Media I for
24 hours. RNA was then isolated as described above. Denatured RNA samples (20 μg) were resolved in a 1% agarose/formaldehyde gel and subsequently transferred onto a nylon membrane (BrightStar-Plus) by capillary diffusion. Blots were prehybridized in 5X Denhardt's solution, 5X saline-sodium phosphate-EDT A, 50%) formamide, 1% SDS, and 100 μg/ml of denatured salmon sperm DNA for 4 hours at 40°C. Subsequently, blots were hybridized with 32P-labeled Cx43 (Beyer et al, 1987) or 18S cDNA (Ambion) overnight at 40°C. The blots were washed 30 min each with 2X SSC (40°C), IX SSC (40°C), and 0.1X SSC with 0.1% SDS (42°C) and then exposed to Kodak Bio-Max film at -80°C for 6-48 hours using intensifying screens.
Data Analysis
Procedures were performed on four or more culture preparations from individual litters of mice. Densitometric analysis of immunoblots, Northern blots and RT-PCR samples were performed using Scion Image software (Scion corporation). Data was presented as means ± SEM. Comparisons between means were analysed using one way analysis of variance with the Tukey's Comparisons test. A P value of less than 0.05 was considered significant.
Mature Astrocytes Express CNTF but not CNTFRα In Vitro
The expression of CNTF and its receptor, CNTFRα, by cortical astrocytes matured in viϊro was compared to that in the cerebral cortex of adult mice. RT-PCR analysis revealed that astrocytes expressed the transcript for CNTF but did not transcribe mRNA for the receptor subunit CNTFRα. When RT-PCR was performed in parallel with RNA isolated from the cerebral cortex, both CNTF and CNTFRα were detected at their expected product sizes. No transcripts were observed when the same RNA samples were amplified in the absence of reverse transcriptase. Thus, astrocytes matured in culture exhibit a CNTF/CNTFRα phenotype comparable to astrocytes in an uninjured or non-reactive state. CNTF Complex Increases Cx43 Protein Expression and Intercellular Coupling
To determine whether CNTF alters Cx43 protein expression in astrocytes, cells were treated with CNTF, CNTFRα, or Complex (equimolar CNTF + CNTFRα) for 3 days and analysed by immunoblotting. Consistent with previous findings (Li and Nagy, 2000; Giaume et al, 1991), the astrocytes matured in viiro were found to express Cx43. However, neither CNTF nor CNTFRα alone altered total Cx43 protein levels when compared to vehicle, while Complex increased total Cx43 by 68 ± 11 %. The phosphorylated states of Cx43 migrate at different rates through the gel and results in banding on the immunoblot (Musil et al, 1990). When the phosphorylated forms of Cx43 were examined, only Complex induced an increase in phosphorylation of the protein; semi-quantitative analysis of phosphorylated Cx43 revealed that only Complex mediated a significant increase in the protein.
Northern blot analysis was used to examine whether the increase in Cx43 was the result of increased mRNA for Cx43. Analysis of RNA collected following 24 hour treatment with the agents revealed that while neither CNTF nor CNTFRα alone caused a change in Cx43 mRNA levels compared to vehicle, Complex increased Cx43 mRNA levels. When normalized for
RNA loading using levels of 18S mRNA, an increase of 107 ± 35 % in Cx43 mRNA over that of vehicle treatment was induced by Complex.
Localization of the Cx43 protein was examined by immunocytochemistry following three-day treatment of the astrocytes with vehicle, CNTF, CNTFRα or Complex. The protein distributed intracellularly as well as to the periphery of the cells. When compared to vehicle treatment, no noticeable difference in either immunostaining intensity or localization of the Cx43 was detected in cells treated with either CNTF or CNTFRα. However, astrocytes treated with Complex showed a dramatic increase in Cx43 immunolabeling. This Complex-induced increase in Cx43 could be detected both within the cytoplasm and at the periphery of the cells. Intercellular coupling between the astrocytes was examined by the scrape-loading/dye transfer technique following three-day treatment of the cells with vehicle, CNTF, CNTFRα or Complex. While neither CNTF or CNTFRα caused an increase in dye transfer between cells within the time period compared to vehicle, Complex increased spreading of the gap junction permeable dye carboxyfluorescein. When quantified from the scrape to the furthest cell exhibiting fluorescence, the distance over which carboxyfluorescein was transferred was significantly greater when cells were treated with Complex, compared to vehicle, CNTF or CNTFRα alone. Complex-induced Increase in Cx43 Expression is Mediated by the JAK/STAT Pathway
Two known intracellular signaling pathways activated by CNTF in other cell systems are the MAPK/ERK pathway and the JAK/STAT pathway (reviewed by Monville et al, 2001). When the matured astrocytes were treated with vehicle, CNTF, CNTFRα, or Complex for 5, 15 and 60 minutes, only Complex caused a detectable increase in the phosphorylation of ERK1 and ERK2 at all time points. While phosphorylation of ERK1/2 was detectable at the 5 minute time-point, Complex induced greater phosphorylation after longer treatments. CNTF alone increased phosphorylation of ERKl and ERK2 only after 60 minute treatment. Previous studies have shown that JAK1 , JAK2 and TYK2 can phosphorylate STAT3 following addition of CNTF (Stahl et al, 1994; Stahl et al, 1995). In this Example, phosphorylation of STAT3 was induced in the astrocytes by both CNTF and Complex treatments and activation of STAT3 was most dramatic at the 5 minute time-point and decreased with longer durations. To determine the pathway mediating the increase in Cx43 by Complex, inhibitors of the
MAPK/ERK pathway and the JAK/STAT pathway were employed. Astrocytes were treated with the ERK-activating kinase (MEK)l/2 inhibitor l,4-diamino-2,3-dicyano-l,4bis[2- aminophenylthio]butadiene (U0126) for 45 minutes prior to and throughout the addition of Complex. Phosphorylation of the ERK1/2 normally induced by Complex was inhibited by U0126. In parallel procedures, astrocytes were pretreated with the JAK2/JAK3 inhibitor α- cyano-(3,4-dihydroxy)-N-benzylcinnamide (AG490). Unlike JAK2, JAK3 expression is limited to hematopoietic and lymphoid cells (Kawamura et al, 1994; Rane and Reddy, 1994) and has not been shown to be associated with CNTF signaling (Stahl et al, 1994). In this Example, AG490 inhibited most, but not all, phosphorylation of STAT3 upon addition of Complex. When RT-PCR analysis for Cx43 mRNA was performed on astrocytes treated with vehicle or Complex in the presence or absence of the pathway inhibitors, Complex was still capable of inducing an increase in Cx43 mRNA levels in the presence of the inhibitor solvent (DMSO) and in the presence of U0126. However, the Complex-induced increase in Cx43 mRNA was not observed in cells pretreated with AG490. Although both CNTF and Complex induce phosphorylation of STAT3 protein, only
Complex causes an upregulation of Cx43. Immunocytochemical analysis of cells treated with CNTF or Complex for 15 minutes revealed a difference in cellular localization of the phosphorylated STAT3. In cells treated with CNTF alone, nuclear translocation was markedly limited resulting in the majority of phosphorylated STAT3 remaining cytosolic. However, when treated with Complex, the majority of phosphorylated STAT3 translocated to the nucleus within the same exposure time.
Cx30 Levels in Mature Cultured Astrocytes are Not Affected by Complex
In addition to Cx43, astrocytes in adult brain or matured in culture express other connexins, including Cx30 (Dahl et al, 1996; Kunzelmann et al, 1999; Nagy et al, 1999). To determine whether Complex altered Cx30 levels, mRNA isolated from astrocytes treated with vehicle, CNTF, CNTFRα, or Complex was analysed by RT-PCR. No change in Cx3 transcript levels could be detected following 24 hour treatment with the agents examined.
Semiquantitative analysis of the Cx30 levels (normalized to GAPDH expression) revealed no significant differences between any of the treatments.
Summary In this Example, the astrocytes cultured from neonatal mice were matured in vilro for seven weeks prior to the described procedures the described procedures. These astrocytes have a mature, non-reactive phenotype, including expression of Cx30 (Dahl et al, 1996; Nagy et al, 1999; Dermietzel et al, 2000; Ip et al, 1993a; Kordower et al, 1997; Maclennan et al, 1996; Monville et al, 2001; Squinto et al, 1990). Previous studies demonstrated that astrocytes exhibit inconsistent or highly variable responses to CNTF. Different states of reactivity exist for astrocytes both in vivo and in vitro, from normal to reactive. Brain disturbances, and in vitro culturing methods are contributing factors to the astrocyte condition (reviewed by Wu and Schwartz, 1998). Reactive astrocytes are known to express both mRNA and protein for CNTFRα (Duberley et al, 1995; Duberley and Johnson, 1996; Ip et al, 1993b; Lee et al, 1997; Rudge et al, 1994a; Rudge et al, 1995). Astrocytes isolated and cultured from neonatal animals and subsequently maintained in culture for a long duration are in a non-reactive state.
In this Example mature murine cortical astrocytes are shown to express CNTF but not CNTFRα. Only when CNTF was administered in the presence of CNTFRα was upregulation of both Cx43 mRNA and protein observed. The regulation of Cx43 expression by Complex was dependent on the JAK/STAT pathway but not the MAPK/ERK pathway. The ability of Complex to upregulate Cx expression is selective as the expression of Cx30 is not affected. Although an increase in Cx43 was detected within the cytoplasm, Complex induced an increase in Cx43 mainly at the periphery of the cells. At the plasma membrane, Cx43 becomes phosphorylated, forms a connexon, which subsequently combines with the connexon from an adjacent cell to form the gap junction (reviewed by Lampe and Lau, 2000; Musil and Goodenough, 1991). Although astrocytes are typically highly coupled, Complex further increased or stabilized functional intercellular communication via gap junction formation. Complex also increases Cx43 and gap junctional coupling in C6 cells as shown in Example A. Therefore, upregulation of Cx43 and intercellular communication by Complex occurs in both gap junction-competent and -deficient cells of the CNS. Upregulation of Cx43 and gap junctions by Complex will circumvent extensive tissue damage in brain injuries, insults and in progressive CNS diseases. Although CNTF has shown limited neuroprotective effects within the CNS following various insults (Kumon et al., 1996; Ogata et al., 1996; Unoki and LaVail, 1994; Wen et al., 1995b), the limitation now appears to be due to the lack of CNTFRα on astrocytes.
EXAMPLE C Gap Junctional Coupling and Neurotoxicity Materials
Dulbecco's Modified Eagle Medium (DMEM), fetal bovine serum (FBS), DMEM/F12, Neurobasal Medium, penicillin-streptomyosin, Hank's balanced salt solution (HBSS) without CaCl2, Phosphate Buffered Saline (PBS), and Earle's Balanced Salt Solution were obtained from GIBCO Laboratories (Burlington, ON, Canada). Cytosine arabinoside, poly-D-lysine, D- glucose, insulin, transferrin, putrescine-HCl, progesterone, sodium selenite, CBX, GZA, AGA, propidium iodide, Hoechst 33342 dye, glutamate, anti-microtubual associated protein 2 (MAP2) antibody, anti-glial fibrillary acidic protein (GFAP) antibody and lactate dehydrogenase (LDH) Detection kit were products of Sigma (Oakville, ON, Canada). L- [3H]glutamate ([3H]Glu, 38-46 Ci/mmol) was purchased from Amersham Canada (Oakville, ON, Canada). Apoptosis Detection System (terminal dUTP nick end labeling, TUNEL, Fluroescein) was from Promega (Madison, WI, USA). Cell strainers (pore size 70 μm) and 6- well plates were acquired from NWR (Mississauga, ON, Canada). Dil (l,l'-dioctadecyl- 3,3,3',3'-tetramethylindocarbocyaine perchlorate) and calcein acetoxymethyl ester (calcein- AM) were purchased from Molecular Probes (Eugene, OR, USA). Solution I consisted of HBSS supplemented with 10%> FBS. Media I was comprised of Neurobasal Medium and DMEM/F12 (2:3) supplemented with 10% FBS. Media II was prepared with 54 ml Neurobasal Medium, 36 ml DMEM/F12, 50 μl penicillin-streptomyosin, and 2 ml of N2 Supplements. N2 Supplements (Mazzoni, I.E., et al, 1991) consisted of the following (given in final concentrations in Media II): glucose (0.6%), insulin (10 μg/ml), transferrin (20 μg/ml), putrescine-HCl (62 μM), progesterone (20 nM), and sodium selenite (30 nM). Sodium Hepes Buffer contained (final concentrations) 134 mM NaCl, 5.2 mM KC1, 1.8 mM CaCl2, 0.8 mM MgSO4, 10 mM glucose, and 20 mM HEPES, pH 7.3.
Cell Culture Primary cultures of type 1 astrocytes were prepared from murine cortices using the method described for Example B. Cortices were dissected from 1 to 2 day old mouse pups, placed into PBS, and subsequently freed of meninges. Cortices were then placed into DMEM supplemented with 10% FBS and were mechanically dissociated using a serological pipette. This cell suspension was then passed through a cell strainer and diluted using DMEM supplemented with 10% FBS and penicillin-streptomyosin at a ratio of 7 ml/cortex. Cells were plated onto 6-well plates (previously coated with poly-D-lysine, 100 μg/ml) in 2 ml aliquots. Cultures were maintained within a humidifed incubator at 37°C in 95% air/5% CO2. Culture media was continuously replaced every three days in addition to shaking the cultures. Astrocytic cultures were maintained for 15-17 days from initial plating prior to use or co- culturing with neurons. Immunocytochemical characterization of cell types present in the final astrocytic cultures revealed that 95-97% of the population were GFAP immunopositive.
Neurons were prepared from mouse cortices in a modified method by Mazzoni and Kenigsberg (1991). Cortices were dissected from embryonic mice of gestation age 16 days and placed into HBSS supplemented with 10% FBS. Cortices were then freed of meninges, minced with scalpels and mechanically disrupted by gently triturating through a fire-polished pasteur pipette (bore size of approximately 0.6 mm internal diameter). Cell suspension was passed through a cell strainer, centrifuged, and subsequently resuspended in HBSS and Media 1 (1:1) supplemented with 10% FBS. Cells (1 x 106 cells/well) were seeded on top of astrocyte cultures and allowed to settle for 2 hours. Subsequently, media was replaced with 2 ml Media II. On day 4, growth media was replaced with fresh Media II containing cytosine arabinoside (2 μM). Thereafter, two-thirds of the conditioned media was replaced every third day with fresh Media II and cultures were maintained for 14 days prior to procedures. Immunocytochemical analysis indicated that cells cultured on top of the astrocytes expressed the neuronal marker MAP2.
Co-cultures consisted of a monolayer of astrocytes to which cortical neurons were seeded on top.
Pre-Loading
Blocking of astrocytic gap junctions was determined by dye coupling using the preloading method as described by Goldberg et al. (1995). Astrocyte cultures were maintained in Media II for 7 days. Narious concentrations of the blocking agents CBX and AGA and the inactive analogue GZA were tested; all agents were added to media 1 hour prior to pre-loading and were present in all solutions. Cultures were bathed in dye solution (5 μM calcein-AM and 10 μM Dil in an isotonic (0.3 M) glucose solution for 20 min in a humidified incubator (37°C, 5% CO2/95% air), rinsed twice with isotonic glucose solution, trypsinized and suspended in DMEM with 1% FBS, and plated onto unlabeled cells at 1:500 ratio of labeled to unlabeled cells. Cells were maintained for 3 hours in the incubator and were subsequently examined with a photomicroscope (Zeiss Axiophot; Carl Zeiss, ΝY, USA). Gap junctional communication was assessed by the passage of calcein from the donor cells (labeled with both calcein and Dil) to the underlying recipient cells.
Glutamate Exposure
Blocking agents were added to co-cultures 1 hour prior and were present in all solutions throughout the procedure. Co-cultures were rinsed with PBS and then bathed in EBSS with glutamate (1 mM) or sham (vehicle) for 3 hours in a humidified incubator (37°C, 95% air/5% CO2). Cells were subsequently maintained for 24 hours post glutamate/sham exposure in fresh Media II.
Mortality Assessment
Samples of the 24 hour conditioned media were collected and analyzed for LDH activity. Cell cultures were exposed to propidium iodide (30 μM in PBS) for 5 minutes and were subsequently fixed in 4% formaldehyde solution. Cells were then stained with TUΝEL and Hoechst and examined by fluorescence microscopy. [3H]Glutamate Uptake
Glutamate uptake procedures were performed in a manner similar to that described by Sitar et al. (1999). Confluent cultures of astrocytes were switched to Media II for 1 week prior to procedures. Blocking agents were added to cultures 1 hour prior to glutamate uptake and were present in all solutions during transport. Cells were rinsed with Sodium Hepes Buffer and exposed to glutamate solution for 120 seconds. Uptake was terminated by washing cultures with ice-cold Tris-sucrose (pH 7.3) solution. Cells were harvested by osmotic lysis (1 ml water/dish) and mechanical scraping. The radioactive contents of the buffer and cells were measured by liquid scintillation counting. Glutamate uptake was computed based on the specific activity of radiolabeled glutamate in the buffer and taken-up by the cells.
Data Analysis
Procedures were performed on three or more culture preparations from individual litters of mice. Data was presented as means ± standard error of the mean (SEM). Comparisons between mean values were evaluated using one way analysis of variance with Student-
Newman-Keuls Multiple Comparisons test. A p value of <0.05 was considered significant.
Blockage of Gap Junctions by CBX and AGA
A minimal concentration of CBX and AGA required to block astrocytic gap junctions was determined by assaying unlabeled cells receiving calcein from a labeled cell (Dil and calcein). Final concentrations of 0 (vehicle), 1, 10, 25 and 50 μM of blockers were tested. While vehicle (water) and GZA had no affect on the passage of calcein from donor cells to, and among, recipient cells, concentrations including and exceeding 25 μM CBX effectively blocked the passage of this dye. A lower concentration of AGA (10 μM) was required to similarly block astrocytic gap junctions while its vehicle (DMSO) had no effect. From the results obtained in pre-loading, concentrations of 25 μM CBX and 10 μM AGA were chosen for use in the remainder of this Example.
Blockage of Gap Junctions Increases Cytotoxicity to Glutamate Glutamate cytotoxicity was determined with primary co-cultures of murine cortical astrocytes and neurons. Immunocytochemical analysis revealed MAP -2 -positive neurons situated on top of a confluent layer of GFAP -positive astrocytes. The two cell types occupied two different focal planes which hindered phase-contrast images but, in conjunction with Hoechst dye staining, allowed for cell-type identification based on nuclear size and its plane of focus.
Glutamate cytotoxicity was analyzed by three mechanisms: LDH release, the inability to exclude propidium iodide, and TUNEL labeling. When presented with a sham insult, neither vehicle, CBX nor GZA caused the cells to release a significant amount of LDH. When glutamate was administered in the presence of vehicle or GZA, no significant LDH release occurred. However, when gap junctions were blocked with CBX, a similar glutamate insult caused the cells to release a significant amount of LDH.
Analysis of propidium iodide labeled cells revealed a similar pattern between agents compared to LDH released. While no difference was ascertained between vehicle, CBX and GZA under sham conditions, glutamate caused a significant increase in propidium iodide- labeled cells in the presence of vehicle and CBX. This glutamate insult, in conjunction with blocked gap junctions (by CBX), caused a substantial number of cells to be labeled with propidium iodide (compared to all other treatments). A third method of cytotoxicity analysis was performed on the co-cultures following glutamate exposure with/without blocking gap junctions using TUNEL. Similar to the other two analyses, neither vehicle, CBX nor GZA alone caused a large proportion of labeling. However, while glutamate exposure induced cytotoxicity (comparing vehicle sham with vehicle glutamate insult), the magnitude of this insult was enhanced when the gap junctions were blocked (glutamate insult with CBX).
To confirm that increased glutamate cytotoxicity by the presence of CBX was caused by blocked gap junctions, another blocker, AGA, was employed. Neither vehicle (DMSO) nor AGA caused a significant LDH release. However, AGA did cause a significant amount of LDH release when present during the glutamate exposure.
Gap Junction Blocker Does Not Affect Astrocytic Glutamate Transporters
As glutamate cytotoxicity was amplified in the presence of a gap junction blocker, it was determined whether the blocker was affecting astrocytic glutamate transporters. There was no difference between the uptake of [3H] glutamate by cortical astrocytes in the presence of vehicle, CBX or GZA. Summary
This Example demonstrates the important role of intercellular communication that allows cells to endure high levels of extracellular glutamate. Glutamate can induce a cytotoxic effect to sensitive cells (i.e. neurons) by one of two mechanisms. In the first mechanism, glutamate activates ionotrophic receptors which subsequently depolarizes the cell resulting in cell swelling and, potentially, necrosis. In the second mechanism, glutamate metabotrophic receptor overstimulation may cause the cell to undergo transcriptionally active suicide (apoptosis). To quantify cell death, we examined markers for both necrosis (LDH release and the inability to exclude PI) and apoptosis (TUNEL labelling).
While the glutamate exposure did not significantly affect LDH release in this Example, PI and TUNEL staining indicated that the dosage of glutamate employed significantly insulted the cells. More specifically, the PI and TUNEL labelling was highly associated with the glutamate-sensitive neurons and labelled very few underlying astrocytes (similar results to Choi (1987)). Astrocytic tolerance to the glutamate insult is likely due to astrocytes lacking NMDA receptors and housing large glycogen stores (a crucial metabolite for energy production).
Although glutamate alone induced cell death, this detrimental effect was amplified in the presence of gap junction blockers, as demonstrated by released LDH, PI- and TUNEL- labelling. Again, mortality markers (PI and TUNEL) were specific to the glutamate-sensitive neurons.
The increased glutamate cytotoxicity in the presence of the gap junction blockers seems directly related to the blockers impeding intercellular gap junctional communication. The blockers alone caused no significant cell death and no significant difference in mortality was determined between vehicle and the inactive blocker in the presence of glutamate.
Furthermore, the blockers did not affect astrocytic uptake of radiolabelled glutamate. The increased mortality seen with the blockers in combination with glutamate could not be attributed to the blockers inhibiting normal astrocytic glutamate transport and sequestering. Thus, neurological diseases that have diminished or compromised gap junctions which increase neuronal vulnerability may be treated by upregulating functional gap junctions. EXAMPLE D Decrease Cell Death by Glutamate Excitotoxicity Cell Culture
Primary co-cultures of murine cortical astrocytes and neurons were prepared as described in the preceding Examples. Astrocyte cultures were established from cortices of 1- to 2-day-old CD-I mouse pups. The cortices were removed from pups, freed of meninges and mechanically dissociated in DMEM (supplemented with 10% FBS; Invitrogen Corp.) using a serological pipette. The cell suspension was passed through a cell strainer (70 μm; NWR) and diluted using DMEM (supplemented with 10%) FBS, 10 units/ml penicillin, and 10 μg/ml streptomycin; Invitrogen Corp.) at a ratio of 7 ml/cortex. Cells were plated onto poly-D-lysine (50 μg/ml) coated 6-well plates in 2 ml aliquots. Cultures were maintained within a humidified incubator at 37°C in 95% air/5% CO2 and media was continuously replaced every 3 days in addition to shaking the cultures. Following a 6 week maturity period, cortical neurons were seeded on top of the astrocyte cultures. Neurons were prepared from cortices of embryonic day 16 CD-I mice. The cortices were freed of meninges, minced using scalpels, and mechanically dissociated by gentle trituration through a fire-polished Pasteur pipette (bore size of approximately 0.6 mm internal diameter). The cell suspension was passed through a cell strainer, centrifuged at 5000 xg for 5 minutes, and resuspended in Hank's Balanced Salt Solution (Invitrogen Corp.) and Media I (Neurobasal Medium and DMEM/F12 (2:3); Invitrogen Corp.) in a 1 :1 ratio supplemented with 10% FBS. Cells (1 x 106 cells/well) were seeded on top of the astrocytes, allowed to settle for 2 hours and, subsequently, maintained in Media II (54 ml Neurobasal Medium, 36 ml DMEM/F12, 50 μl penicillin-streptomycin, glucose (0.6%), insulin (10 μg/ml), transferrin (20 μg/ml), putrescine-HCl (62 μM), progesterone (20 nM), and sodium selenite (30 nM)). On day 4, a fresh media change containing cytosine arabinoside (2 μM) was performed. Thereafter, two-thirds of the conditioned media was replaced with fresh Media II every 3 days. Co-cultures were maintained for 2 weeks prior to procedures.
Immunocytochemistry Co-cultures were rinsed with phosphate buffered saline (PBS) and fixed with 4% formaldehyde (in PBS) for 10 minutes. Non-specific binding of antibodies was blocked by incubating the cultures with 10% normal goat serum (in PBS). Cultures were then incubated with microtubule associated protein 2 (MAP2) monoclonal antibody and glial fibrillary acidic protein (GFAP) polyclonal antibody (Sigma). Following three washes with PBS, cultures were incubated in Alexa- and fluorescein-conjugated secondary IgG (Molecular Probes) for 1 hour and then mounted with Nectashield mounting medium (Nector). Immunostaining was viewed using a Zeiss Axioskop microscope and images were captured using Northern Exposure, Nersion2 (ImageExperts Inc.).
Exposure to Agents and Glutamate
Prior to glutamate insult, co-cultures were treated with vehicle (PBS), CΝTF (20 ng/ml; R&D Systems), soluble CΝTFRα (200 ng/ml; R&D Systems), or Complex (20 ng/ml CΝTF + 200 ng/ml CΝTFRα) with a fresh media change every 24 hours for 72 hours. Twenty-four hours following the final pretreatment, the gap junction blocker CBX (25 μM; Sigma), its inactive analogue GZA (25 μM; Sigma), or solvent (H2O) was added to the co-cultures for one hour. These compounds were present in all solutions thereafter. Co-cultures were rinsed with PBS and then bathed in Earle's Balanced Salt Solution (Invitrogen Corp.) with glutamate (1 mM) or PBS (sham insult) for 3 hours in a humidified incubator (37°C, 95% air/5% CO2). Following the insult, cells were maintained in fresh Media II.
Injury Assessment
Twenty-four hours following glutamate exposure, media samples were collected from the co-cultures, centrifuged at 5000 xg for 5 minutes, stored at -20°C, and later examined for lactate dehydrogenase (LDH) content using the CytoTox 96 Νon-Radioactive Cytotoxicity Assay (Promega). The co-cultures were bathed in propidium iodide solution (30 μM in PBS) for 5 minutes, rinsed three times with PBS (5 minutes each), and fixed with 4% formaldehyde (in PBS). The fixed co-cultures were stained with terminal dUTP nick end labelling (TUΝEL; DeadEnd Fluorometric TUΝEL System; Promega) and with DAPI nucleic acid stain
(Molecular Probes). The stained cells were rinsed three times with PBS and mounted using Nectashield mounting medium (Nector). Staining was viewed and images were captured as outlined above.
Data Analysis
Procedures were performed on four or more culture preparations from individual litters of mice. Data was presented as means ± standard error of the mean (SEM). Comparisons between means were analysed using one way analysis of variance with the Tukey's Comparisons test. A P value of less than 0.05 was considered significant.
Pretreatment of Co-cultures with Complex Decreases Glutamate Cytotoxicity Immunocytochemical analysis revealed that the neurons were established superficial to the astrocyte monolayer. Neurons were distinctly identified from the astrocytes based on expression of MAP2, lack of GFAP expression, smaller nuclei, and occupation of a separate focal plane when examined by microscopy. No cytotoxic effects, as revealed by LDH release, propidium iodide staining, and TUNEL labelling, were identified in the co-cultures exposed to CNTF, CNTFRα, Complex, GZA, or CBX. Furthermore, no differences in cytotoxicity were detected between GZA and solvent (H2O) treatments under all conditions tested. In addition, exposure of 1 mM glutamate to the co-cultures induced significant cytotoxicity, as determined by the three markers of cell death examined.
Pretreatment of the co-cultures with CNTF or CNTFRα resulted in LDH release similar to that with vehicle pretreatment. However, glutamate exposure did not cause a significant release of LDH by the cells when co-cultures were pretreated with Complex and was significantly less than that released with vehicle and CNTFRα pretreatments
While the number of cells stained with propidium iodide in the co-cultures following glutamate exposure was significantly increased within vehicle and CNTFRα pretreatment groups, no significant change was seen with CNTF and Complex pretreatments. Furthermore, the number of propidium iodide cells was significantly reduced following glutamate insult with Complex pretreatment as compared to vehicle or CNTFRα pretreatments.
Glutamate exposure caused a significant insult in the co-cultures as determined by TUNEL-labelling (vehicle pretreatment). Similar glutamate-induced cytotoxicity as marked by TUNEL-labelling was seen in CNTFRα-pretreated co-cultures. However, considerably less number of cells stained with TUNEL was observed in the co-cultures pretreated with CNTF and Complex. Comparisons between groups revealed that only Complex pretreatment significantly reduced the number of TUNEL-positive cells, as compared to vehicle pretreatment.
Glutamate-induced Cytotoxicity Increase When Gap Junctions Are Blocked
A significant increase in LDH release occurred in co-cultures pretreated with vehicle, CNTF, CNTFRα and Complex following glutamate exposure in the presence of CBX. When comparing between pretreatment groups, only pretreatment of co-cultures with Complex caused a significant decreased in LDH release following glutamate exposure with CBX, as compared to vehicle pretreatment.
The presence of CBX caused a significant increase in propidium idodide-positive cells following glutamate exposure within all pretreatment groups. However, when compared to vehicle pretreatment, only Complex pretreatment significantly reduced the quantity of propidium idodide-positive cells following glutamate exposure with CBX.
When gap junctions were blocked with CBX, a significant increased the number of TUNEL-positive cells occurred following glutamate exposure under all pretreatment conditions. When comparing between pretreatment groups, both CNTF and Complex pretreatments significantly reduced the number of TUNEL-labelled cells in the glutamate- insulted co-cultures with CBX, as compared to vehicle pretreatment under similar conditions.
Summary Heightened expression of Cx43 and gap junctional coupling in the neuro-supportive astrocytes, extends the neuroprotective properties of these cells. Treatment of neuronal and astrocytic co-cultures with Complex to upregulate both Cx43 expression and functional gap junctional coupling reduces cytotoxicity following a glutamate insult. Markers of both necrosis (LDH release and propidium idodide staining) and apoptosis (TUNEL labelling) are increased following the glutamate insult. The majority of propidium idodide staining and TUNEL- labelling is limited to the smaller neurons located superficial to the astrocytes.
Glutamate-induced cytotoxicity in neuron and astrocyte co-cultures is reduced by CNTF pretreatment. Since neurons express CNTFRα, CNTF alone can directly activate those cells and potentially increase their resilience to glutamate excitotoxicity. Skaper et al. (1992) but the extended neuroprotection shown in this Example requires administration of both CNTF and CNTFRα.
Although CNTF causes a reduction in glutamate-induced cytotoxicity within its own pretreatment group, this difference is not significantly different from vehicle pretreatment under similar conditions. When co-cultures were pretreated with CTNF in combination with CNTFRα, a substantial reduction in the glutamate-induced cytotoxicity was observed.
Complex pretreatment not only prevented cytotoxicity within its own pretreatment group, but also significantly reduced cytotoxicity when compared to vehicle pretreatment with a similar insult. The detrimental effect of glutamate cytotoxicity is amplified when gap junctions are blocked with CBX. However, pretreatment of the co-cultures with Complex in this Example caused a significant reduction in the glutamate-induced cytotoxicity in the presence of CBX, as compared to vehicle pretreatment. This shows that the neuroprotective effect of Complex is due to both gap junction-dependent and gap junction -independent actions.
EXAMPLE E Rescuing brain tissue from induced stroke in mice by Complex injection
To determine the ability of Complex to reduce neuronal death in vivo, the "Complex" of CNTF-(soluble)CNTFRα was administered to mice following induced stroke. Stroke was induced in mice by middle cerebral artery occlusion (MCAO). Occlusion of the blood vessel in mice is a widely used stroke model (Siushansian et al., 2001 ; Nakase et al., 2003). Immediately following the insult (2 hour occlusion in the mouse model), the respective agents (vehicle, CNTF, or Complex) were administered via intraperitoneal injection. Since CNTF administration into both rodents and humans induces weight reduction, the body weights and food intake (provided ad libum) were closely monitored. Following twice daily injections of the agents for 4 days post-injury, the animals were euthanized and the brain sectioned and examined for Cx43 expression as well as for markers of brain injury. Extent of injury was assessed by two methods: first, infarct (core and penumbra) volume of the brain was measured following staining the sections with thionin; second, the sections were labeled with TUNEL (terminal dUTP nick end labeling) and immunoreacted against caspase3, two markers used to identify programmed cell death.
The results, in a study using 2 mice/agent, confirmed that the effects of soluble CNTFRα bound to CNTF does not hinder the dimeric molecule from inducing CNTF-like physiological effects. Within 4 days post stroke, mice injected with Complex mimicked body weight reduction to that of CNTF-injected mice. A large portion of the body weight reduction can be attributed to the agents inducing loss of both appetite and thirst since food and H2O intake were significantly reduced compared to vehicle. Furthermore, the loss in appetite is solely attributed to the effect of Complex and independent of the stroke since Complex injection without stroke caused similar results to that of Complex with insult.
The initial concentration of Complex chosen for injections to reduce stroke-induced brain damage was that in which the same concentration of CNTF alone would induce physiological changes (i.e. weight loss; Fantuzzi et al., 1995). As with other agents used to circumvent stroke, such as the blood thinner warfarin, excessive administration of Complex may cause secondary side-effects which override the agent from reducing brain tissue damage. Therefore, the concentration of Complex may be titered to determine the amount most effective for neuroprotection in vivo, by for example reducing brain tissue damage without causing substantial side-effects.
To complement the requirement of both CNTF and CNTFRα in reduction of brain injury following stroke, MCAO insults are performed on knockout mice. While CNTFRα knockout mice are not viable, mice lacking CNTF expression thrive normally. Using mice in which the CNTF gene has been deleted through homologous recombination (generous gift of Dr. Micheal Sendtner, Universitaat Wϋurzburg), the importance of CNTF expression in circumventing brain injury following stroke is assessed. A similar MCAO insult and similar methods to assess brain injury as outlined herein is performed on the CNTF knockout mice and compared to wild-type littermates. Furthermore, in cases where the lack of endogenous CNTF expression leads to increased brain injury in the knockout mice, rescue of brain injury by administration of exogenous CNTF or Complex following the insult is determined.
EXAMPLE F
Identification of Additional Genes Involved in Neuroprotection
Further characterization of the effect of Complex on gene expression in nerve tissue may be used to identify specific genes that are up- or downregulated which contribute to the neuroprotection to that above and beyond CNTF treatment alone. Furthermore, identification of specific genes whose expression is modified by Complex treatment permits selective up- or downregulation of those genes and subsequently enhancement of neuroprotection.
To identify differential gene expression affected by Complex, co-cultures of neurons and astrocytes, and pure neuronal cultures (i.e. primary cortical neurons and/or NT2 cells), are treated with vehicle, CNTF or Complex, and subsequently processed to isolate total RNA which will be analyzed by, for example, gene arrays. Narious genes are differentially expressed by treatment of cultures with Complex compared to vehicle or CΝTF treatment. The identified differential gene candidates are confirmed by other techniques (e.g. northern blot analysis and/or reverse transcription polymerase chain reactions).
REFERENCES
The following publications are incorporated by reference. Patents and patent applications: U.S. 4,997,929, U.S. 5,011,914, U.S. 5,426,177, U.S. 5,892,003, U.S. 10/073/658 (June 13, 2002), WO 91/04316.
Adler R, Landa KB, Manthorpe M, and Naron S (1979) Cholinergic neuronotrophic factors: intraocular distribution of trophic activity for ciliary neurons. Science 204:1434-1436.
Alvarez-Maubecin N, Garcia-Hernandez F, Williams JT, and Nan Bockstaele EJ (2000)
Functional coupling between neurons and glia. JNeurosci 20:4091-4098. Arakawa Y, Sendtner M, and Thoenen H (1990) Survival effect of ciliary neurotrophic factor (CΝTF) on chick embryonic motoneurons in culture: comparison with other neurotrophic factors and cytokines. JNeurosci 10:3507-3515.
Asada H, Ip ΝY, Pan L, Razack Ν, Parfitt MM, and Plunkett RJ (1995) Time course of ciliary neurotrophic factor mRΝA expression is coincident with the presence of protoplasmic astrocytes in traumatized rat striatum. JNeurosci Res 40:22-30. Asada H, Kaseloo PA, Lis A, Petti DM, and Plunkett RJ (1996) Traumatized rat striatum produces neurite-promoting and neurotrophic activities in vitro . Exp Neurol 139:173-
187. Bani-Yaghoub M, Bechberger JF, Underhill TM, Νaus CC. The effects of gap junction blockage on neuronal differentiation of human ΝTera2/clone DI cells. Exp Neurol 1999;156:16-32 Barbin G, Manthorpe M, and Naron S (1984) Purification of the chick eye ciliary neuronotrophic factor. JNeurochem 43:1468-1478. Barker FG, Chang SM, Gutin PH, Malec MK, McDermott MW, Prados MD, and Wilson CB (1998) Survival and functional status after resection of recurrent glioblastoma multiforme. Neurosurgery 42:709-720. Baumann, H, S.F. Ziegler, B. Mosley, K.K. Morella, S. Pajovic, and D.P. Gearing. 1993.
Reconstitution of the response to leukemia inhibitory factor, oncostatin M, and ciliary neurotrophic factor in hepatoma cells. J. Biol. Chem. 268:8414-8417. Beck T, Lindholm D, Castren E, and Wree A (1994) Brain-derived neurotrophic factor protects against ischemic cell damage in rat hippocampus. J Cereb Blood Flow Metab 14:689- 692.
Benda P, Lightbody J, Sato G, Levine L, and Sweet W (1968) Differentiated rat glial cell strain in tissue culture. Science 161:370-371. Bennett MN and Goodenough DA (1978) Gap junctions, electrotonic coupling, and intercellular communication. Neurosci Res Program Bull 16:1 -486. Bennett MN, Barrio LC, Bargiello TA, Spray DC, Hertzberg E, Saez JC. Gap junctions: new tools, new answers, new questions. Neuron 1991;6:305-320 Bergles DE, Jahr CE. Synaptic activation of glutamate transporters in hippocampal astrocytes.
Neuron 1997;19:1297-1308 Beyer, E.G., D.L. Paul, and D.A. Goodenough. 1987. Connexin43: a protein from rat heart homologous to a gap junction protein from liver. J Cell Biol 105:2621-2629. Blanc EM, Bruce-Keller AJ, and Mattson MP (1998) Astrocytic gap junctional communication decreases neuronal vulnerability to oxidative stress-induced disruption of Ca2+ homeostasis and cell death. J Neurochem 70:958-970. Blottner D, Bruggemann W, and Unsicker K (1989) Ciliary neurotrophic factor supports target- deprived preganglionic sympathetic spinal cord neurons. Neurosci Lett 105:316-320. Brosnan CF, Scemes E, and Spray DC (2001) Cytokine regulation of gap junction connectivity: an open-and-shut case or changing partners at the Nexus? Am J Paihol
158:1565-1569. Bruzzone R, Ressot C. Connexins, gap junctions and cell-cell signalling in the nervous system.
Eur J Neurosci 1997;9:1-6 Cedarbaum JM (corresponding author)(1995) The pharmacokinetics of subcutaneously administered recombinant human ciliary neurotrophic factor (rHCNTF) in patients with amyotrophic lateral sclerosis: relation to parameters of the acute-phase response. The
ALS CNTF Treatment Study (ACTS) Phase I-II Study Group. Clin Neuropharmacol
18:500-514. Chan KM, Lam DT, Pong K, Widmer HR, and Hefti F (1996) Neurotroρhin-4/5 treatment reduces infarct size in rats with middle cerebral artery occlusion. Neurochem Res
21:763-767. Cheung RT and Cechetto DF (1997) Colchicine affects cortical and amygdalar neurochemical changes differentially after middle cerebral artery occlusion in rats. J Comp Neurol
387:27-41. Choi DW (1988) Glutamate neurotoxicity and diseases of the nervous system. Neuron 1 :623-
634. Choi DW, Maulucci-Gedde M, Kriegstein AR. Glutamate neurotoxicity in cortical cell culture.
J Neurosci 1987;7:357-368 Clatterbuck RE, Price DL, and Koliatsos NE (1993) Ciliary neurotrophic factor prevents retrograde neuronal death in the adult central nervous system. Proc Nail Acad Sci USA
90:2222-2226. Cornell-Bell AH, Finkbeiner SM, Cooper MS, Smith SJ. Glutamate induces calcium waves in cultured astrocytes: long-range glial signaling. Science 1990;247:470-473
Cotrina ML, Kang J, Lin JH et al. Astrocytic gap junctions remain open during ischemic conditions. J Neurosci 1998;18:2520-2537 Coyle JT and Schwarcz R (1976) Lesion of striatal neurones with kainic acid provides a model for Huntington's chorea. Naϊure 263:244-246. Dahl, E., D. Manthey, Y. Chen, H.J. Schwarz, Y.S. Chang, P.A. LaUey, B.J. Nicholson, and K.
Willecke. 1996. Molecular cloning and functional expression of mouse connexin-30,a gap junction gene highly expressed in adult brain and skin. J. Biol. Chem. 271:17903-
17910. Davidson JS, Baumgarten IM, Harley EH. Reversible inhibition of intercellular junctional communication by glycyrrhetinic acid. Biochem Biophys Res Commun 1986; 134:29-
36 Davidson JS, Baumgarten IM. Glycyrrhetinic acid derivatives: a novel class of inhibitors of gap-junctional intercellular communication. Structure-activity relationships. J
Pharmacol Exp Ther 1988;246:1104-1107 Davis S, Aldrich TH, Ip NY, Stahl N, Scherer S, Farruggella T, DiStefano PS, Curtis R,
Panayotatos N, and Gascan H (1993) Released form of CNTF receptor alpha component as a soluble mediator of CNTF responses. Science 259:1736-1739. Davis S, Aldrich TH, Valenzuela DM, Wong W, Furth ME, Squinto SP, and Yancopoulos
GD (1991) The receptor for ciliary neurotrophic factor. Science 253:59-63. Davis, S., T.H. Aldrich, N. Stahl, L. Pan, T. Taga, T. Kishimoto, N.Y. Ip, and G.D.
Yancopoulos. 1993b. LIFR beta and gpl30 as heterodimerizing signal transducers of the tripartite CNTF receptor. Science 260:1805-1808. Deck JH, Eng LF, Bigbee J, and Woodcock SM (1978) The role of glial fibrillary acidic protein in the diagnosis of central nervous system tumors. Ada Neuropathol(Berl) 42:183-190.
Del Bigio MR, Yan HJ, and Xue M (2001) Intracerebral infusion of a second-generation ciliary neurotrophic factor reduces neuronal loss in rat striatum following experimental intracerebral hemorrhage. J Neurol Sci 192:53-59. Dermietzel R and Spray DC (1993) Gap junctions in the brain: where, what type, how many and why? Trends Neurosci 16:186-192. Dermietzel R, Gao Y, Scemes E, et al. Connexin43 null mice reveal that astrocytes express multiple connexins. Brain Res Brain Res Rev 2000;32:45-56 Dermietzel R, Hertberg EL, Kessler JA, and Spray DC (1991) Gap junctions between cultured astrocytes: immunocytochemical, molecular, and electrophysiological analysis. J
Neurosci 11:1421-1432. Dhein S (1998) Gap junction channels in the cardiovascular system: pharmacological and physiological modulation. Trends Pharmacol Sci 19:229-241. Duberley RM, Johnson IP, Anand P, Swash M, Martin J, Leigh PN, and Zeman S (1995)
Ciliary neurotrophic factor receptor expression in spinal cord and motor cortex in amyotrophic lateral sclerosis. J Neurol Sci 129 Suppl: 109-113. Duberley, R.M. and I.P. Johnson. 1996. Increased expression of the alpha subunit of the ciliary neurotrophic factor (CNTF) receptor by rat racial motoneurons after neonatal axotomy and CNTF treatment. Neurosci Lett 218:188-192. el Fouly, M.H., J.E. Trosko, and C.C. Chang. 1987. Scrape-loading and dye transfer. A rapid and simple technique to study gap junctional intercellular communication. Exp. Cell
Res. 168:422-430. Enkvist MO and McCarthy KD (1994) Astro glial gap junction communication is increased by treatment with either glutamate or high K+ concentration. J Neurochem 62:489-495.
Fantuzzi G, Benigni F, Sironi M, Conni M, Carelli M, Cantoni L, Shapiro L, Dinarello CA,
Sipe JD, and Ghezzi P (1995) Ciliary neurotrophic factor (CNTF) induces serum amyloid A, hypoglycaemia and anorexia, and potentiates IL-1 induced corticosterone and IL-6 production in mice. Cytokine 7:150-156. Favata, M.F., K.Y. Horiuchi, E.J. Manos, A.J. Daulerio, D.A. Stradley, W.S. Feeser, D.E. Van
Dyk, W.J. Pitts, R.A. Earl, F. Hobbs, R.A. Copeland, R.L. Magolda, P.A. Scherle, and
J.M. Trzaskos. 1998. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J. Biol. Chem. 273:18623-18632. Fedoroff S, Richardson A. Generation of Mouse Astroglia and Microglia Cultures from Mouse Neopallium; in Fedoroff S, Richardson A (eds): Protocols for Neuronal Cell Cultures.
Totowa, New Jersey, Humana Press, 1997, pp 131-141 Formigli L, Papucci L, Tani A et al. Aponecrosis: morphological and biochemical exploration of a syncretic process of cell death sharing apoptosis and necrosis. J Cell Physiol 2000;182:41-49 Froes MM, Campos de Carvalho AC. Gap junction-mediated loops of neuronal-glial interactions. Glia 1998;24:97-107
Froes MM, Correia AH, Garcia- Abreu J, Spray DC, Campos de Carvalho AC, Neto MN. Gap- junctional coupling between neurons and astrocytes in primary central nervous system cultures. Proc Νatl Acad Sci U S A 1999;96:7541-7546 Gardner-Medwin AR. Analysis of potassium dynamics in mammalian brain tissue. J Physiol (Lond) 1983;335:393-426
Gearing, D.P., S.F. Ziegler, M.R. Comeau, D. Friend, B. Thoma, D. Cosman, L. Park, and B. Mosley. 1994. Proliferative responses and binding properties of hematopoietic cells transfected with low-affinity receptors for leukemia inhibitory factor, oncostatin M, and ciliary neurotrophic factor. Proc. Natl. Acad. Sci. U. S. A 91 :1119-1123. Giaume C, Fromaget C, el Aoumari A, Cordier J, Glowinski J, and Gros D (1991) Gap junctions in cultured astrocytes: single-channel currents and characterization of channel- forming protein. Neuron 6:133-143. Giaume, C. and K.D. McCarthy. 1996. Control of gap-junctional communication in astrocytic networks. Trends Neurosci 19:319-325. Gilula ΝB, Reeves OR, and Steinbach A (1972) Metabolic coupling, ionic coupling and cell contacts. Nature 235:262-265. Gluckman P, Klempt Ν, Guan J, Mallard C, Sirimanne E, Dragunow M, Klempt M, Singh K, Williams C, and Νikolics K (1992) A role for IGF-1 in the rescue of CΝS neurons following hypoxic-ischemic injury. Biochem Biophys Res Commun 182:593-599. Goldberg GS, Bechberger JF, and Νaus CC (1995) A pre-loading method of evaluating gap junctional communication by fluorescent dye transfer [published erratum appears in Biotechniques 1995 Aug;19(2):212]. Biotechniques 18:490-497. Goldberg GS, Lampe PD, Nicholson BJ. Selective transfer of endogenous metabolites through gap junctions composed of different connexins. Nat Cell Biol 1999;1:457-459 Goodenough DA (1976) In vitro formation of gap junction vesicles. J Cell Biol 68:220-231. Grasl-Kraupp B, Ruttkay-Nedecky B, Koudelka H, Buckowska K, Bursch W, Schulte-
Hermann R. In situ detection of fragmented DNA (TUNEL assay) fails to discriminate among apoptosis, necrosis, and autolytic cell death: a cautionary note. Hepatology
1995;21:1465-1468 Hagg T and Naron S (1993) Ciliary neurotrophic factor prevents degeneration of adult rat substantia nigra dopaminergic neurons in vivo. Proc Natl Acad Sci U S A 90:6315- 6319.
Hagg T, Quon D, Higaki J, and Naron S (1992) Ciliary neurotrophic factor prevents neuronal degeneration and promotes low affinity ΝGF receptor expression in the adult rat CΝS.
Neuron 8:145-158. Hatten M and Mason C (1986) Νeuron-astroglial interactions in vitro and in vivo. Trends Neurosci 9:168-174.
Hattori, R., Ν. Maulik, H. Otani, L. Zhu, G. Cordis, R.M. Engelman, M.A. Siddiqui, and D.K.
Das. 2001. Role of STAT3 in ischemic preconditioning. J. Mol. Cell Cardiol. 33:1929-
1936. Henderson JT, Seniuk ΝA, Richardson PM, Gauldie J, and Roder JC (1994) Systemic administration of ciliary neurotrophic factor induces cachexia in rodents. J Clin Invest
93:2632-2638. Henrich-Νoack P, Prehn JH, and Krieglstein J (1996) TGF-beta 1 protects hippocampal neurons against degeneration caused by transient global ischemia. Dose-response relationship and potential neuroprotective mechanisms. Stroke 27:1609-1614. Hertz L. Possible role of neuroglia: a potassium-mediated neuronal—neuroglial—neuronal impulse transmission system. Nature 1965;206:1091-1094 Hirano T, Nakajima K, and Hibi M (1997) Signaling mechanisms through gpl30: a model of the cytokine system. Cytokine Growth Factor Rev 8:241-252. Hodge, D.R., D. Li, S.M. Qi, and W.L. Farrar. 2002. IL-6 induces expression of the Fli-1 proto-oncogene via STAT3. Biochem. Biophys. Res. Commun. 292:287-291.
Hossain MZ, Peeling J, Sutherland GR, Hertzberg EL, and Nagy JI (1994) Ischemia-induced cellular redistribution of the astrocytic gap junctional protein connexin43 in rat brain.
Brain Res 652:311-322. Huang RP, Fan Y, Hossain MZ, Peng A, Zeng ZL, and Boynton AL (1998) Reversion of the neoplastic phenotype of human glioblastoma cells by connexin 43 (cx43). Cancer Res
58:5089-5096. Huang RP, Hossain MZ, Sehgal A, and Boynton AL (1999) Reduced connexin43 expression in high-grade human brain glioma cells. JSurg Oncol 70:21-24. Ip NY, Li YP, van dS, I, Panayotatos N, Alderson RF, and Lindsay RM (1991) Ciliary neurotrophic factor enhances neuronal survival in embryonic rat hippocampal cultures.
JNeurosci 11:3124-3134. Ip NY, McClain J, Barrezueta NX, Aldrich TH, Pan L, Li Y, Wiegand SJ, Friedman B, Davis S, and Yancopoulos GD (1993a) The alpha component of the CNTF receptor is required for signaling and defines potential CNTF targets in the adult and during development. Neuron 10:89-102. Ip NY, Wiegand SJ, Morse J, and Rudge JS (1993) Injury-induced regulation of ciliary neurotrophic factor mRNA in the adult rat brain. Eur JNeurosci 5:25-33. Ip, N.Y. and G.D. Yancopoulos. 1992. Ciliary neurotrophic factor and its receptor complex.
Prog Growth Factor Res 4:139-155. Iwata F, Joh T, Ueda F, Yokoyama Y, Itoh M. Role of gap junctions in inhibiting ischemia- reperfusion injury of rat gastric mucosa. Am J Physiol 1998;275:G883-G888 John, G.R., E. Scemes, S.O. Suadicani, J.S. Liu, P.C. Charles, S.C. Lee, D.C. Spray, and CF. Brosnan. 1999. IL-lbeta differentially regulates calcium wave propagation between primary human fetal astrocytes via pathways involving P2 receptors and gap junction channels. Proc Natl Acad Sci U S A 96 : 11613 - 11618. Kahn MA, Ellison JA, Chang RP, Speight GJ, and de Vellis J (1997) CNTF induces GFAP in a
S-100 alpha brain cell population: the pattern of CNTF-alpha R suggests an indirect mode of action. Brain Res Dev Brain Res 98 :221 -233.
Kandler K, Katz LC. Neuronal coupling and uncoupling in the developing nervous system.
Curr Opin Neurobiol 1995;5:98-105 Kawamura, M., D.W. McVicar, J.A. Johnston, T.B Blake, Y.Q. Chen, B.K. Lai, A.R. Lloyd,
D.J. Kelvin, J.E. Staples, J.R. Ortaldo, and J.L. O'Shea. 1994. Molecular cloning of L- JAK, a Janus family protein-tyrosine kinase expressed in natural killer cells and activated leukocytes. Proc Natl Acad Sci USA 91:6374-6378. Kelloff GJ, Crowell JA, Boone CW et al. Clinical development plan: 18beta-glycyrrhetinic acid. J Cell Biochem Suppl 1994;20:166-175 Kermer P, Klocker N, and Bahr M (1999) Neuronal death after brain injury. Models, mechanisms, and therapeutic strategies in vivo. Cell Tissue Res 298:383-395.
Kirino T, Tsujita Y, and Tamura A (1991) Induced tolerance to ischemia in gerbil hippocampal neurons. J Cereb Blood Flow Metab 11 :299-307. Kitagawa K, Matsumoto M, Tagaya M, Hata R, Ueda H, Niinobe M, Handa N, Fukunaga R, Kimura K, Mikoshiba K (1990) 'Ischemic tolerance' phenomenon found in the brain. Brain Res 528:21-24.
Kobayashi, H. & Mizisin, A.P. (2000) Neuropeptides 34: 338-47 Koizumi J. Glycogen in the central nervous system. Prog Histochem Cytochem 1974;6:1-37
Koketsu N, Berlove DJ, Moskowitz MA, Kowall NW, Caday CG, and Finklestein SP (1994) Pretreatment with intraventricular basic fibroblast growth factor decreases infarct size following focal cerebral ischemia in rats. Ann Neurol 35:451-457.
Kordower, J.H., C. Yaping, and A.J. Maclennan. 1997. Ciliary neurotrophic factor receptor alpha-immunoreactivity in the monkey central nervous system. J Comp Neurol
377:365-380.
Kumar NM and Gilula NB (1996) The gap junction communication channel. Cell 84:381-388.
Kumon, Y., S. Sakaki, H. Watanabe, K. Nakano, S. Ohta, S. Matsuda, H. Yoshimura, and M. Sakanaka. 1996. Ciliary neurotrophic factor attenuates spatial cognition impairment, cortical infarction and thalamic degeneration in spontaneously hypertensive rats with focal cerebral ischemia. Neurosci Lett 206:141-144.
Kunzelmann, P., W. Schroder, O. Traub, C. Steinhauser, R. Dermietzel, and K. Willecke.
1999. Late onset and increasing expression of the gap junction protein connexin30 in adult murine brain and long-term cultured astrocytes. Glia 25:111-119. Lambert PD, Anderson KD, Sleeman MW, Wong V, Tan J, Hijarunguru A, Corcoran TL,
Murray JD, Thabet KE, Yancopoulos GD, and Wiegand SJ (2001) Ciliary neurotrophic factor activates leptin-like pathways and reduces body fat, without cachexia or rebound weight gain, even in leptin-resistant obesity. Proc Nail Acad Sci USA 98:4652-4657.
Lampe PD and Lau AF (2000) Regulation of gap junctions by phosphorylation of connexins. Arch Biochem Biophys 384:205-215.
Lee MY, Deller T, Kirsch M, Frotscher M, and Hofinann HD (1997) Differential regulation of ciliary neurotrophic factor (CNTF) and CNTF receptor alpha expression in astrocytes and neurons of the fascia dentata after entorhinal cortex lesion. JNeurosci 17 :1137- 1146. Li, W.E. and J.I. Nagy. 2000. Connexin43 phosphorylation state and intercellular communication in cultured astrocytes following hypoxia and protein phosphatase inhibition. Eur. J. Neurosci. 12:2644-2650. Lin LF, Mismer D, Lile JD, Amies LG, Butler ET, Nannice JL, and Collins F (1989)
Purification, cloning, and expression of ciliary neurotrophic factor (CΝTF). Science
246:1023-1025. Lin TΝ, Wang PY, Chi SI, and Kuo JS (1998) Differential regulation of ciliary neurotrophic factor (CΝTF) and CΝTF receptor alpha (CΝTFR alpha) expression following focal cerebral ischemia. Brain Res Mol Brain Res 55:71-80. Loewenstein WR (1981) Junctional intercellular communication: the cell-to-cell membrane channel. Physiol Rev 61 : 829-913. Loewenstein WR and Kanno Y (1966) Intercellular communication and the control of tissue growth: lack of communication between cancer cells. Naiure 209:1248-1249.
Lucas DR and Newhouse JP (1957) The toxic effect of sodium L-glutamate on the inner layers of the retina. Arch Ophihalmol 58:193-201. Ludolph AC, Meyer T, and Riepe MW (2000) The role of excitotoxicity in ALS-what is the evidence? J Neurol 247 Suppl 1:17-16. Ma J, Qiu J, Hirt L, Dalkara T, and Moskowitz MA (2001) Synergistic protective effect of caspase inhibitors and bFGF against brain injury induced by transient focal ischaemia.
BrJ Pharmacol 133:345-350. Maclennan AJ, Ninson EΝ, Marks L, McLaurin DL, Pfeifer M, and Lee Ν (1996)
Immunohistochemical localization of ciliary neurotrophic factor receptor alpha expression in the rat nervous system. J Neurosci 16:621-630.
Malgrange B, Rogister B, Lefebvre PP, Mazy-Servais C, Welcher AA, Bonnet C, Hsu RY,
Rigo JM, Nan De Water TR, and Moonen G (1998) Expression of growth factors and their receptors in the postnatal rat cochlea. Neurochem Res 23:1133-1138. Martin W, Zempel G, Hulser D, Willecke K. Growth inhibition of oncogene-transformed rat fibroblasts by cocultured normal cells: relevance of metabolic cooperation mediated by gap junctions. Cancer Res 1991;51:5348-5351 Martinou JC, Martinou I, and Kato AC (1992) Cholinergic differentiation factor (CDF/LIF) promotes survival of isolated rat embryonic motoneurons in vitro. Neuron 8:737-744. Masiakowski P, Liu HX, Radziejewski C, Lottspeich F, Oberthuer W, Wong N, Lindsay RM, Furth ME, and Panayotatos Ν (1991) Recombinant human and rat ciliary neurotrophic factors. J Neurochem 57:1003-1012. Masu Y, Wolf E, Holtmann B, Sendtner M, Brem G, and Thoenen H (1993) Disruption of the
CΝTF gene results in motor neuron degeneration. Naiure 365:27-32. Mazzoni IE, Kenigsberg RL. Thrombin indirectly affects cholinergic cell expression in primary septal cell cultures in a manner distinct from nerve growth factor. Neuroscience
1991;45:195-204 McGeer EG and McGeer PL (1976) Duplication of biochemical changes of Huntington's chorea by intrastriatal inj ections of glutamic and kainic acids . Naiure 263:517-519.
McNeill H, Williams C, Guan J, Dragunow M, Lawlor P, Sirimanne E, Nikolics K, and
Gluckman P (1994) Neuronal rescue with transforming growth factor-beta 1 after hypoxic-ischaemic brain injury. Neuroreport 5:901-904. Micevych PE and Abelson L (1991) Distribution of mRNAs coding for liver and heart gap junction proteins in the rat central nervous system. J Comp Neurol 305:96-118.
Micevych PE, Popper P, and Hatton GI (1996) Connexin 32 mRNA levels in the rat supraoptic nucleus: up-regulation prior to parturition and during lactation. Neuroendocrinology
63:39-45. Monville, C, M. Coulpier, L. Conti, C. De-Fraja, P. Dreyfus, C. Fages, D. Riche, M. Tardy, E. Cattaneo, and M. Peschanski. 2001. Ciliary neurotrophic factor may activate mature astrocytes via binding with the leukemia inhibitory factor receptor. Mol Cell Neurosci
17:373-384. Morales R, Duncan D. Specialized contacts of astrocytes with astrocytes and with other cell types in the spinal cord of the cat. Anat Rec 1975;182:255-265 Muller W, Afra D, and Schroder R (1977) Supratentorial recurrences of gliomas.
Morphological studies in relation to time intervals with astrocytomas. Acta Neurochir Wien) 37:75-91. Murry CE, Jennings RB, and Reimer KA (1986) Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation 74:1124-1136. Musil, L.S., B.A. Cunningham, G.M. Edelman, and D.A. Goodenough. 1990. Differential phosphorylation of the gap junction protein connexin43 in junctional communication- competent and -deficient cell lines. J. Cell Biol. 111:2077-2088. Musil,L. S. and D.A. Goodenough. 1991. Biochemical analysis of connexin43 intracellular transport, phosphorylation, and assembly into gap junctional plaques. J. Cell Bio 115:1357-1374.
Nadarajah B, Jones AM, Evans WH, and Parnavelas JG (1997) Differential expression of connexins during neocortical development and neuronal circuit formation. JNeurosci
17:3096-3111. Nadarajah B, Thomaidou D, Evans WH, and Parnavelas JG (1996) Gap junctions in the adult cerebral cortex: regional differences in their distribution and cellular expression of connexins. J Comp Neurol 376:326-342. Nadler JN, Perry BW, and Cotman CW (1978) Intraventricular kainic acid preferentially destroys hippocampal pyramidal cells. Naiure 271 :676-677.
Νagy, J.I., D. Patel, P.A. Ochalski, and G.L. Stelmack. 1999. Connexin30 in rodent, cat and human brain: selective expression in gray matter astrocytes, co-localization with connexin43 at gap junctions and late developmental appearance. Neuroscience 88:447-
468. Νakata Ν, Kato H, and Kogure K (1993) Protective effects of basic fibroblast growth factor against hippocampal neuronal damage following cerebral ischemia in the gerbil. Brain
Res 605:354-356. Νaus CC, Bechberger JF, Caveney S, and Wilson JX (1991) Expression of gap junction genes in astrocytes and C6 glioma cells. Neurosci Lett 126:33-36. Νaus CC, EHsevich K, Zhu D, BeHiveau D J, and Del Maestro RF (1992) In vivo growth of C6 glioma cells transfected with connexin43 cDΝA. Cancer Res 52:4208-4213. Νaus CC, Ozog MA, Bechberger JF, and Νakase T (2001) A neuroprotective role for gap junctions. CellAdhes Commun 8:325-328. Νaus, CC, J.F. Bechberger, Y. Zhang, L. Nenance, H. Yamasaki, S.C Juneja, G.M .Kidder, and C. Giaume. 1997. Altered gap junctional communication, intercellular signaling, and growth in cultured astrocytes deficient in connexin43. J. Neurosci. Res. 49:528-
540. Nedergaard M (1994) Direct signaling from astrocytes to neurons in cultures of mammalian brain cells. Science 263:1768-1771. Nedergaard M, Cooper AJ, Goldman SA. Gap junctions are required for the propagation of spreading depression. J Neurobiol 1995;28:433-444 Negro, A., G. Corona, E. Bigon, I. Martini, C Grandi, S.D. Skaper, and L. Callegaro. 1991.
Synthesis, purification, and characterization of human ciliary neuronotrophic factor from E. coli. JNeurosci Res 29:251-260. Nesbitt, J.E., N.L. Fuentes, and G.M. Fuller. 1993. Ciliary neurotrophic factor regulates fibrinogen gene expression in hepatocytes by binding to the interleukin-6 receptor.
Biochem. Biophys. Res. Commun. 190:544-550. Nieto-Sampedro M, Lewis ER, Cotman CW, Manthorpe M, Skaper SD, Barbin G, Longo FM, and Naron S (1982) Brain injury causes a time-dependent increase in neuronotrophic activity at the lesion site. Science 217:860-861. Νishimune, H, S. Nasseur, S. Wiese, M.C Birling, B. Holtmann, M. Sendtner, J.L. Iovanna, and CE. Henderson. 2000. Reg-2 is a motoneuron neurotrophic factor and a signalling intermediate in the CΝTF survival pathway. Nal Cell Biol 2:906-914. Νozaki K, Finklestein SP, and Beal MF (1993) Basic fibroblast growth factor protects against hypoxia-ischemia and ΝMDA neurotoxicity in neonatal rats. J Cereb Blood Flow
Melab 13:221-228. Ochalski PA, Sawchuk MA, Hertzberg EL, Νagy, JI. Astrocytic gap junction removal, connexin43 redistribution, and epitope masking at excitatory amino acid lesion sites in rat brain. Glia 1995;14:279-294 Ogata Ν, Ogata K, Imhof HG, and Yonekawa Y (1996) Effect of CΝTF on ischaemic cell damage in rat hippocampus. Ada Neurochir (Wien) 138:580-583. Olney JW and Sharpe LG (1969) Brain lesions in an infant rhesus monkey treated with monsodium glutamate. Science 166:386-388. Oppenheim RW, Prevette D, Yin QW, Collins F, and MacDonald J (1991) Control of embryonic motoneuron survival in vivo by ciliary neurotrophic factor. Science
251:1616-1618. Ozog MA, Siushansian R, and Νaus CC (2002) Blocked gap junctional coupling increases glutamate-induced neurotoxicity in neuron-astrocyte co-cultures. J Neuropaihol Exp
Neurol 61:132-141. Park CK, Ju WK, Hofmann HD, Kirsch M, Ki KJ, Chun MH, and Lee MY (2000) Differential regulation of ciliary neurotrophic factor and its receptor in the rat hippocampus following transient global ischemia. Brain Res 861 :345-353.
Parpura N, Basarsky TA, Liu F, Jeftinija K, Jeftinija S, and Haydon PG (1994) Glutamate- mediated astrocyte-neuron signalling. Naiure 369:744-747. Patterson, P.H. 1992. The emerging neuropoietic cytokine family: first CDF/LIF, CΝTF and
IL-6; next OΝC, MGF, GCSF? Curr Opin Neurobiol 2:94-97. Perez-Pinzon MA, Nitro TM, Dietrich WD, and Sick TJ (1999) The effect of rapid preconditioning on the microglial, astrocytic and neuronal consequences of global cerebral ischemia. Ada Neuropathol (Bed) 97:495-501. Perez-Pinzon MA, Xu GP, Dietrich WD, Rosenthal M, and Sick TJ (1997) Rapid preconditioning protects rats against ischemic neuronal damage after 3 but not 7 days of reperfusion following global cerebral ischemia. J Cereb Blood Flow Metab 17:175-182. Phelps CH. An ultrastructural study of methionine sulphoximine-induced glycogen accumulation in astrocytes of the mouse cerebral cortex. J Neurocytol 1975;4:479-490
Rane, S.G. and E.P. Reddy. 1994. JAK3: a novel JAK kinase associated with terminal differentiation of hematopoietic cells. Oncogene 2415-2423. Regan RF, Choi DW. Excitotoxicity and central nervous system trauma; in Salzman SK, Faden
AI (eds): The Neurobiology of Central Nervous System Trauma. New York, Oxford University Press, 1994, pp 173-181
Reuss, B., M. Hertel, S. Werner, and K. Unsicker. 2000. Fibroblast growth factors-5 and -9 distinctly regulate expression and function of the gap junction protein connexin43 in cultured astroglial cells from different brain regions. Glia 30:231-241. Reuss, B., R. Dermietzel, and K. Unsicker. 1998. Fibroblast growth factor 2 (FGF-2) differentially regulates connexin (ex) 43 expression and function in astroglial cells from distinct brain regions. Glia 22:19-30. Robe PA, Rogister B, Merville MP, and Bours V (2000) Growth regulation of astrocytes and
C6 cells by TGFbetal : correlation with gap junctions. Neuroreport 11 :2837-2841. Robinson MB, Dowd LA. Heterogeneity and functional properties of subtypes of sodium- dependent glutamate transporters in the mammalian central nervous system. Adv
Pharmacol 1997;37:69-115 Roser K, Bohn W, Giese G, and Mannweiler K (1991) Subclones of C6 rat glioma cells differing in intermediate filament protein expression. Exp Cell Res 197:200-206. Rothman S. Synaptic release of excitatory amino acid neurotransmitter mediates anoxic neuronal death. J Neurosci 1984;4: 1884-1891
Rothman SM and Olney JW (1986) Glutamate and the pathophysiology of hypoxic— ischemic brain damage. Ann Neurol 19:105-111. Rothman SM and Olney JW (1995) Excitotoxicity and the NMDA receptor— still lethal after eight years. Trends Neurosci 18:57-58. Rothstein JD, Dykes-Hoberg M, Pardo CA, et al. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron
1996;16:675-686 Rouach, N. and C Giaume. Connexins and gap junctional communiacation in astrocytes are targets for neuroglial interaction. 203-214. 2001. Ref Type: Generic Rozental R, Giaume C, Spray DC. Gap junctions in the nervous system. Brain Res Brain Res
Rev 2000;32:11-15 Rozental R, Srinivas M, and Spray DC (2001) How to close a gap junction channel. Efficacies and potencies of uncoupling agents. Melhods Mol Biol 154:447-476. Rozental, R., C Giaume, and D.C. Spray. 2000. Gap junctions in the nervous system. Brain
Res. Brain Res. Rev. 32:11-15. Rudge JS, Alderson RF, Pasnikowski EM, McClain J, Ip NY, and Lindsay RM (1992) Expression of Ciliary Neurotrophic Factor and the Neurotrophins - Nerve Growth
Factor, Brain-Derived Neurotrophic Factor and Neurotrophin 3 - in Cultured Rat
Hippocampal Astrocytes. Eur JNeurosci 4:459-471. Rudge JS, Li Y, Pasnikowski EM,
Mattsson K, Pan L, Yancopoulos GD, Wiegand SJ, Lindsay RM, and Ip NY (1994b)
Neurotrophic factor receptors and their signal transduction capabilities in rat astrocytes. Eur JNeurosci 6:693-705.
Rudge JS, Morrissey D, Lindsay RM, and Pasnikowski EM (1994a) Regulation of ciliary neurotrophic factor in cultured rat hippocampal astrocytes. Eur JNeurosci 6:218-229. Rudge, J.S., E.M. Pasnikowski, P. Hoist, and R.M. Lindsay. 1995. Changes in neurotrophic factor expression and receptor activation following exposure of hippocampal neuron astrocyte cocultures to kainic acid. JNeurosci 15:6856-6867.
Saggio I, Gloaguen I, Poiana G, and Laufer R (1995) CNTF variants with increased biological potency and receptor selectivity define a functional site of receptor interaction. EMBO J
14:3045-3054. Salcman M (1995) Glioblastoma and malignant astrocytoma, in Brain iumors (Kaye A and Laws Jr E eds) pp 449-477, Churchill Livingstone, Hong Kong.
Sambrook J, Fritsch EF, and Maniatis T (1989) Extraction, Purification, analysis of messenger
RNA from eukaryotic cells, in Molecular Cloning: A Laboraϊory Manual Cold Springs
Harbor Laboratories, Cold Spring, NY. Sasaki K, Oomura Y, Suzuki K, Hanai K, and Yagi H (1992) Acidic fibroblast growth factor prevents death of hippocampal CA1 pyramidal cells following ischemia. Neurochem Inl
21:397-402. Schabitz WR, Schwab S, Spranger M, and Hacke W (1997) Intraventricular brain-derived neurotrophic factor reduces infarct size after focal cerebral ischemia in rats. J Cereb
Blood Flow Melab 17:500-506. Sendtner M, Kreutzberg GW, and Thoenen H (1990) Ciliary neurotrophic factor prevents the degeneration of motor neurons after axotomy. Naiure 345:440-441.
Sendtner M, Schmalbruch H, Stockli KA, Carroll P, Kreutzberg GW, and Thoenen H (1992)
Ciliary neurotrophic factor prevents degeneration of motor neurons in mouse mutant progressive motor neuronopathy. Naiure 358:502-504. Shigeno T, Mima T, Takakura K, Graham DI, Kato G, Hashimoto Y, and Furukawa S (1991) Amelioration of delayed neuronal death in the hippocampus by nerve growth factor. J
Neurosci 11:2914-2919. Simon RP, Swan JH, Griffiths T, and Meldrum BS (1984) Blockade of N-methyl-D-aspartate receptors may protect against ischemic damage in the brain. Science 226:850-852. Simpson I, Rose B, and Loewenstein WR (1977) Size limit of molecules permeating the junctional membrane channels. Science 195:294-296.
Sitar SM, Hanifi-Moghaddam P, Gelb A, Cechetto DF, Siushansian R, Wilson JX. Propofol prevents peroxide-induced inhibition of glutamate transport in cultured astrocytes.
Anesthesiology 1999;90:1446-1453 Siushansian R, Bechberger JF, Cechetto DF, Hachinski VC, and Naus CC (2001) Connexin43 null mutation increases infarct size after stroke. J Comp Neurol 440:387-394.
SiuYi, L.D., K. Unsicker, and B. Reuss. 2001. Gap junctions modulate survival-promoting effects of fibroblast growth factor-2 on cultured midbrain dopaminergic neurons. Mol.
Cell Neurosci. 18 :44-55. Skaper SD, Negro A, Dal Toso R, and Facci L (1992) Recombinant human ciliary neurotrophic factor alters the threshold of hippocampal pyramidal neuron sensitivity to excitotoxin damage: synergistic effects of monosialogangliosides. J Neurosci Res 33:330-337. Sloviter RS (1983) "Epileptic" brain damage in rats induced by sustained electrical stimulation of the perforant path. I. Acute electrophysiological and light microscopic studies. Brain
Res Bull 10:675-697. Sohl G, Guldenagel M, Beck H et al. Expression of connexin genes in hippocampus of kainate- treated and kindled rats under conditions of experimental epilepsy. Brain Res Mol
Brain Res 2000;83:44-51 Spray DC, Dermietzel R. Gap junctions in the nervous system: an introduction; in Spray DC, Dermietzel R (eds): Gap Junctions in the Nervous System. Georgetown, R.G. Landes Co., 1996, pp 1-11 Squinto, S.P., T.H. Aldrich, R.M. Lindsay, D.M. Morrissey, N. Panayotatos, S.M. Bianco, M.E. Furth, and G.D. Yancopoulos. 1990. Identification of functional receptors for ciliary neurotrophic factor on neuronal cell lines and primary neurons. Neuron 5:757- 766. Stahl, N., T.G. Boulton, T. Farruggella, N.Y. Ip, S.Davis, B.A. Witthuhn, F.W. Queue, O.
Silvennoinen, G. Barbieri, and S. Pellegrini. 1994. Association and activation of Jak- Tyk kinases by CNTF-LIF-OSM-IL-6 beta receptor components. Science 263 :92-95.
Stahl, N., TJ. Farruggella, T.G. Boulton, Z. Zhong, J.E.J. Darnell, and G.D. Yancopoulos. 1995. Choice of STATs and other substrates specified by modular tyrosine-based motifs in cytokine receptors. Science 267:1349-1353. Stockli KA, Lillien LE, Naher-Noe M, Breitfeld G, Hughes RA, Raff MC, Thoenen H, and Sendtner M (1991) Regional distribution, developmental changes, and cellular localization of CNTF-mRNA and protein in the rat brain. J Cell Biol 115 :447-459. Stockli KA, Lottspeich F, Sendtner M, Masiakowski P, Carroll P, Gotz R, Lindholm D, and
Thoenen H (1989) Molecular cloning, expression and regional distribution of rat ciliary neurotrophic factor. Naiure 342:920-923. Symes, A., S. Lewis, L. Corpus, P. Rajan, S.E. Hyman, and J.S. Fink. 1994. STAT proteins participate in the regulation of the vasoactive intestinal peptide gene by the ciliary neurotrophic factor family of cytokines. Mol Endocrinol 8:1750-1763. Tabernero A, Giaume C, Medina JM. Endothelin-1 regulates glucose utilization in cultured astrocytes by controlling intercellular communication through gap junctions. Glia 1996;16:187-195
Taga T, Hibi M, Hirata Y, Yamasaki K, Yasukawa K, Matsuda T, Hirano T, and Kishimoto T (1989) Interleukin-6 triggers the association of its receptor with a possible signal transducer, gpl30. Cell 58:573-581. Takahashi R, Yokoji H, Misawa H, Hayashi M, Hu J, and Deguchi T (1994) A null mutation in the human CNTF gene is not causally related to neurological diseases. Nat Genet 7:79-
84. Tanaka K, Tsukahara T, Hashimoto N, Ogata N, Yonekawa Y, Kimura T, and Taniguchi T
(1994) Effect of nerve growth factor on delayed neuronal death after cerebral ischaemia. A a Neurochir (Wien) 129:64-71. Tanaka K, Watase K, Manabe T, et al. Epilepsy and exacerbation of brain injury in mice lacking the glutamate transporter GLT-1. Science 1997;276:1699-1702
Teubner B, Odermatt B, Guldenagel M et al. Functional expression of the new gap junction gene connexin47 transcribed in mouse brain and spinal cord neurons. J Neurosci
2001;21:1117-1126 Theriault E, Frankenstein UN, Hertzberg EL, and Nagy JI (1997) Connexin43 and astrocytic gap junctions in the rat spinal cord after acute compression injury. J Comp Neurol
382:199-214. Trosko JE and Ruch RJ (1998) Cell-cell communication in carcinogenesis. Froni Biosci
3:D208-D236. Truettner, J., R. Busto, W. Zhao, M.D. Ginsberg, and M.A. Perez-Pinzon. 2002. Effect of ischemic preconditioning on the expression of putative neuroprotective genes in the rat brain. Brain Res. Mol. Brain Res. 103:106-115. Tsukahara T, Yonekawa Y, Tanaka K, Ohara O, Wantanabe S, Kimura T, Nishijima T, and
Taniguchi T (1994) The role of brain-derived neurotrophic factor in transient forebrain ischemia in the rat brain. Neurosurgery 34:323-331. Tsuzuki N, Miyazawa T, Matsumoto K, Nakamura T, and Shima K (2001) Hepatocyte growth factor reduces the infarct volume after transient focal cerebral ischemia in rats. Neurol
Res 23:417-424. Unoki, K. and M.M. LaNail. 1994. Protection of the rat retina from ischemic injury by brain- derived neurotrophic factor, ciliary neurotrophic factor, and basic fibroblast growth factor. Invesi Ophthalmol Vis Sci 35:907-915.
Nukelic JI, Yamamoto T, Hertzberg EL, Νagy JI. Depletion of connexin43-immunoreactivity in astrocytes after kainic acid-induced lesions in rat brain. Neurosci Lett 1991;130:120-
124 Waelsch H. Glutamic acid and cerebral function. Advances in Protein Chemistry 1951;6:299- 341
Weis, J. et al. (1999) J. Neurooncol. 44:243-53 Wen R, Song Y, Cheng T, Matthes MT, Yasumura D, LaNail MM, and Steinberg RH (1995)
Injury-induced upregulation of bFGF and CΝTF mRΝAS in the rat retina. JNeurosci
15:7377-7385. Wen, T.C, S. Matsuda, H. Yoshimura, T. Kawabe, and M. Sakanaka. 1995b. Ciliary neurotrophic factor prevents ischemia-induced learning disability and neuronal loss in gerbils. Neurosci Lett 191:55-58. Wieloch T (1985) Hypoglycemia-induced neuronal damage prevented by an Ν-methyl-D- aspartate antagonist. Science 230:681-683. Wiese S, Digby MR, Gunnersen JM, Gotz R, Pei G, Holtmann B, Lowenthal J, and Sendtner M (1999) The anti-apoptotic protein ITA is essential for ΝGF-mediated survival of embryonic chick neurons. Nat Neurosci 2:978-983. Willecke K, Eiberger J, Degen J, Eckardt D, Romualdi A, Guldenagel M, Deutsch U, and Sohl
G (2002) Structural and functional diversity of connexin genes in the mouse and human genome. Biol Chem 383:725-737. Wu, N.W. and J.P. Schwartz. 1998. Cell culture models for reactive gliosis: new perspectives.
JNeurosci Res 51 :675-681. Xu B, Dube MG, Kalra PS, Farmerie WG, Kaibara A, Moldawer LL, Martin D, and Kalra SP
(1998) Anorectic effects of the cytokine, ciliary neurotropic factor, are mediated by hypothalamic neuropeptide Y: comparison with leptin. Endocrinology 139:466-473. Yamamoto T, Ochalski A, Hertzberg EL, and Νagy JI (1990) LM and EM immunolocalization of the gap junctional protein connexin 43 in rat brain. Brain Res 508:313-319. Yamasaki H (1990) Gap junctional intercellular communication and carcinogenesis.
Carcinogenesis 11:1051-1058. Yang CW, Lim SW, Han KW, Ahn HJ, Park JH, Kim YH, Kirsh M, Cha JH, Kim YS, Kim J, and Bang BK (2001 ) Upregulation of ciliary neurotrophic factor (CΝTF) and CΝTF receptor alpha in rat kidney with ischemia-reperfusion injury. J Am Soc Nephrol
12:749-757. Zahs KR. Heterotypic coupling between glial cells of the mammalian central nervous system.
Glia 1998;24:85-96 Zhu CZ and Auer RΝ (1994) Intraventricular administration of insulin and IGF-1 in transient forebrain ischemia. J Cereb Blood Flow Meiab 14:237-242. Zhu D, Caveney S, Kidder GM, and Naus CC (1991) Transfection of C6 glioma cells with connexin 43 cDNA: analysis of expression, intercellular coupling, and cell proliferation. Proc Nail Acad Sci USA 88:1883-188.
OTHER EMBODIMENTS
Although various embodiments of the invention are disclosed herein, many adaptations and modifications may be made within the spirit and scope of the invention in accordance with the common general knowledge of those skilled in this art. Such modifications include the substitution of known equivalents for any aspect of the invention in order to achieve the same result in substantially the same way. Accession numbers, as used herein, refer to Accession numbers from multiple databases, including GenBank, the European Molecular Biology Laboratory (EMBL), the DNA Database of Japan (DDBJ), or the Genome Sequence Data Base (GSDB), for nucleotide sequences, and including the Protein Information Resource (PIR), SWISSPROT, Protein Research Foundation (PRF), and Protein Data Bank (PDB) (sequences from solved structures), as well as from translations from annotated coding regions from nucleotide sequences in GenBank, EMBL, DDBJ, or RefSeq, for polypeptide sequences. Numeric ranges are inclusive of the numbers defining the range. In the specification, the word "comprising" is used as an open-ended term, substantially equivalent to the phrase "including, but not limited to", and the word "comprises" has a corresponding meaning. Citation of references herein shall not be construed as an admission that such references are prior art to the present invention. All publications are incorporated herein by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein and as though fully set forth herein. The invention includes all embodiments and variations substantially as hereinbefore described and with reference to the examples and drawings.

Claims

WE CLAIM:
1. A method for providing neuroprotection in a subject in need thereof, comprising administering to the subject effective amounts of: a) a CNTF peptide or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding a CNTF peptide or a biologically-active fragment or variant thereof; and b) a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof.
2. A method of modulating cell degeneration or cell death in a subject in need thereof, comprising administering to the subject effective amounts of: a) a CNTF peptide or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding the CNTF peptide or a biologically-active fragment or variant thereof; and b) a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof.
3. A method of treatment or prophylaxis of a neurodegenerative disease in a subject in need thereof, comprising administering to the subject effective amounts of: a) a CNTF peptide or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding the CNTF peptide or a biologically-active fragment or variant thereof; and b) a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof.
4. A method of ameliorating the cytotoxic effect of a compound in a subject in need thereof, comprising administering to the subject effective amounts of: a) a CNTF peptide or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding the CNTF peptide or a biologically-active fragment or variant thereof; and b) a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof, or a nucleic acid molecule encoding a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof.
5. The method of any one of claims 1 through 4, wherein the subject is suffering from neural insult or injury.
6. The method of any one of claims 1 through 4, wherein the subject is suffering from a condition selected from substance abuse, trauma, stroke, ischemia, Huntington's disease, Alzheimer's disease, Parkinson's disease, prion disease, variant Creutzfeld- Jakob disease, amyotrophic lateral sclerosis (ALS), olivopontocerebellar atrophy, epilepsy, seizures, and hypoglycemic encephalopathy.
7. The method of any one of claims 1 through 4, wherein the subject is undergoing surgery or other intervention.
8. The method of any one of claims 1 -7, wherein the cell degeneration or cell death, neurodegenerative disease, or cytotoxic effect is characterized by glutamate excitotoxicity.
9. The method of any one of claims 1 through 8, wherein the peptide that acts as a receptor for CNTF is soluble.
10. The method of claim 9, wherein the soluble peptide is CNTFRα.
11. The method of any one of claims 1 through 10, wherein the peptides are administered as a complex.
12. The method of any one of claims 1 through 11, wherein the subject is a human.
13. Use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof, for neuroprotection.
14. Use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof, for preparation of a medicament for neuroprotection.
15. Use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof, for modulation of cell degeneration or cell death.
16. Use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof, for preparation of a medicament for modulation of cell degeneration or cell death.
17. Use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof, for treatment or prophylaxis of a neurodegenerative disorder.
18. Use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof, for preparation of a medicament for treatment or prophylaxis of a neurodegenerative disorder.
19. Use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof, for ameliorating the cytotoxic effect of a compound.
20. Use of a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and a peptide that acts as a receptor for CNTF or a biologically- active fragment or variant thereof, for preparation of a medicament for ameliorating the cytotoxic effect of a compound.
21. A composition comprising a CNTF peptide or biologically-active fragment or varient thereof, a peptide that acts as a receptor for CNTF or a biologically-acitve fragment or varient thereof, and a pharmaceutically acceptable carrier for the use of claim 12, 14, 16 or 18.
22. A method of screening for a neuroprotective compound, the method comprising:
(a) providing a first system comprising a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof;
(b) providing a second system comprising a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof;
(c) providing a third system comprising:
(i) a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and
(ii) a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof;
(d) contacting the first and second systems with a test compound; and
(e) determining whether the test compound modulates neuroprotection in the first or second system when compared to the third system.
23. A method of screening for a neuroprotective compound, the method comprising:
(a) providing a system comprising:
(i) a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof, and
(ii) a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof;
(b) contacting the system with a test compound; and
(c) determining whether the test compound modulates neuroprotection in the presence or absence of a gap junction blocker.
24. The method of claim 22 or 23, wherein the neuroprotection comprises protection of neuronal cells or neuronal-associated cells from injury or degeneration.
25. The method of claim 24, wherein the neuroprotection comprises increasing the expression level of a connexin.
26. The method of claim 25, wherein the connexin is Cx43.
27. A method of screening for a nucleic acid molecule that modulates neuroprotection, the method comprising:
(a) providing a neuronal or neuronal-associated cell;
(b) contacting the neuronal or neuronal-associated cell with: (i) a peptide having the activity of a CNTF peptide or a biologically-active fragment or variant thereof;
(ii) a peptide that acts as a receptor for CNTF or a biologically-active fragment or variant thereof; or
(iii) a Complex comprising the CNTF and the peptide that acts as a receptor for CNTF; and
(c) determining the level of nucleic acid molecule expression in (i), (ii), and (iii), wherein a change in the level of nucleic acid molecule expression in (iii), when compared to the level of nucleic acid molecule expression in (i) or (ii) is indicative of a nucleic acid molecule that modulates neuroprotection.
28. The method of claim 27, wherein the determining is done by a microarray.
29. The method of any one of claims 22 through 28, wherein the peptide that acts as a receptor for CNTF is soluble.
30. The method of claim 29, wherein the soluble peptide is CNTFRα.
31. The method of any one of claims 22 through 28, wherein the peptide having the activity of a CNTF peptide is CNTF.
32. The use of any one of claims 13 through 20, wherein the peptide that acts as a receptor for CNTF is soluble.
33. The use of claim 32, wherein the soluble peptide is CNTFRα.
34. The use of any one of claims 13 through 20, wherein the peptide having the activity of a CNTF peptide is CNTF.
PCT/CA2003/001626 2002-10-24 2003-10-24 Methods for neuroprotection WO2004037281A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003275846A AU2003275846A1 (en) 2002-10-24 2003-10-24 Methods for neuroprotection

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US42068102P 2002-10-24 2002-10-24
US60/420,681 2002-10-24

Publications (1)

Publication Number Publication Date
WO2004037281A1 true WO2004037281A1 (en) 2004-05-06

Family

ID=32176610

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2003/001626 WO2004037281A1 (en) 2002-10-24 2003-10-24 Methods for neuroprotection

Country Status (2)

Country Link
AU (1) AU2003275846A1 (en)
WO (1) WO2004037281A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5846935A (en) * 1992-10-09 1998-12-08 Regeneron Pharmaceuticals, Inc. Modified ciliary neurotrophic factors
WO2002022149A1 (en) * 2000-09-14 2002-03-21 Ares Trading S.A. Use of il-6r/il-6 chimera in huntington's disease
WO2002064021A2 (en) * 2001-02-12 2002-08-22 Baylor College Of Medicine Methods and compositions for control of bone formation via modulation of ciliary neurotrophic factor activity

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5846935A (en) * 1992-10-09 1998-12-08 Regeneron Pharmaceuticals, Inc. Modified ciliary neurotrophic factors
US6440702B1 (en) * 1992-10-09 2002-08-27 Regeneron Pharmaceuticals, Inc. Nucleic acids encoding modified ciliary neurotrophic factors
WO2002022149A1 (en) * 2000-09-14 2002-03-21 Ares Trading S.A. Use of il-6r/il-6 chimera in huntington's disease
WO2002064021A2 (en) * 2001-02-12 2002-08-22 Baylor College Of Medicine Methods and compositions for control of bone formation via modulation of ciliary neurotrophic factor activity

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
NAUS C C ET AL: "A neuroprotective role for gap junctions.", CELL COMMUN ADHES, 8 (4-6) 325-8. JOURNAL CODE: 101096596. ISSN: 1541-9061., 2001, XP008026947 *
OZOG MARK A ET AL: "Ciliary neurotrophic factor ( CNTF ) in combination with its soluble receptor (CNTFRalpha) increases connexin43 expression and suppresses growth of C6 glioma cells.", CANCER RESEARCH, (2002 JUN 15) 62 (12) 3544-8. JOURNAL CODE: 2984705R. ISSN: 0008-5472., June 2002 (2002-06-01), XP002232406 *

Also Published As

Publication number Publication date
AU2003275846A1 (en) 2004-05-13

Similar Documents

Publication Publication Date Title
Pennica et al. Cardiotrophin-1, a cytokine present in embryonic muscle, supports long-term survival of spinal motoneurons
Lucius et al. The angiotensin II type 2 (AT2) receptor promotes axonal regeneration in the optic nerve of adult rats
Rosenstein et al. Neurotrophic effects of vascular endothelial growth factor on organotypic cortical explants and primary cortical neurons
Saxena et al. Leptin as a novel profibrogenic cytokine in hepatic stellate cells: mitogenesis and inhibition of apoptosis mediated by extracellular regulated kinase (Erk) and Akt phosphorylation
US7563862B2 (en) Neural regeneration peptides and methods for their use in treatment of brain damage
AU2003273615B2 (en) Neuroprotective synergy of erythropoietin and insulin-like growth factor
US8673594B2 (en) Methods for stimulating nervous system regeneration and repair by regulating arginase I and polyamine synthesis
KR20010085767A (en) Method for detecting, preventing, and treating renal disorders by modulating, regulating, and inhibiting connective tissue growth factor
Honsa et al. Increased expression of hyperpolarization‐activated cyclic nucleotide‐gated (HCN) channels in reactive astrocytes following ischemia
US20100015099A1 (en) Materials and methods relating to treatment of injury and disease to the central nervous system
Rudge et al. Regulation of ciliary neurotrophic factor in cultured rat hippocampal astrocytes
Qu et al. The synapse-associated protein rapsyn regulates tyrosine phosphorylation of proteins colocalized at nicotinic acetylcholine receptor clusters
AU2001259453A1 (en) Methods for stimulating nervous system regeneration and repair by regulating arginase 1 and polyamine synthesis
US7605128B2 (en) Neurogenerative or neurotrophic factors for mitigating a symptom of ischemia
WO2004037281A1 (en) Methods for neuroprotection
US8481486B2 (en) Methods of treating neurological diseases by regulating migration of neuroblasts in the adult nervous system with tenascin-R
WO2003004612A2 (en) Inhibitor of t cell activation
Gorba et al. Neural regeneration protein is a novel chemoattractive and neuronal survival-promoting factor
EP0658117B1 (en) Treatment of tumors with neurotrophic factors and cell proliferation inhibitors
KR20180121046A (en) OCT4 expression or ubiquitination regulating composition comprising CHIP
JP2006525244A (en) Insulin-inducible genes as therapeutic targets for diabetes
Messina et al. Mechanisms of developmental cell death
CN113144170A (en) Peptides and their use in modulating megakaryocyte differentiation and treating platelet-related disorders
WO2007119180A2 (en) Survival and development of neural cells
Nair Synaptic targeting of neurotransmitter receptors is regulated by neurobeachin

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP