WO2001000193A2 - Zinc ionophores as anti-apoptotic agents - Google Patents

Zinc ionophores as anti-apoptotic agents Download PDF

Info

Publication number
WO2001000193A2
WO2001000193A2 PCT/IB2000/001005 IB0001005W WO0100193A2 WO 2001000193 A2 WO2001000193 A2 WO 2001000193A2 IB 0001005 W IB0001005 W IB 0001005W WO 0100193 A2 WO0100193 A2 WO 0100193A2
Authority
WO
WIPO (PCT)
Prior art keywords
zinc
pyrithione
ionophore
group
body weight
Prior art date
Application number
PCT/IB2000/001005
Other languages
French (fr)
Other versions
WO2001000193A3 (en
Inventor
Henry Fliss
Original Assignee
Zinc Therapeutics Canada, Inc.
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 Zinc Therapeutics Canada, Inc. filed Critical Zinc Therapeutics Canada, Inc.
Priority to JP2001505903A priority Critical patent/JP2003503346A/en
Priority to AU18396/01A priority patent/AU1839601A/en
Priority to EP00979231A priority patent/EP1189604A2/en
Priority to CA002375852A priority patent/CA2375852A1/en
Publication of WO2001000193A2 publication Critical patent/WO2001000193A2/en
Publication of WO2001000193A3 publication Critical patent/WO2001000193A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/07Retinol compounds, e.g. vitamin A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/145Amines having sulfur, e.g. thiurams (>N—C(S)—S—C(S)—N< and >N—C(S)—S—S—C(S)—N<), Sulfinylamines (—N=SO), Sulfonylamines (—N=SO2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/27Esters, e.g. nitroglycerine, selenocyanates of carbamic or thiocarbamic acids, meprobamate, carbachol, neostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/315Zinc compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/325Carbamic acids; Thiocarbamic acids; Anhydrides or salts thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • A61K31/355Tocopherols, e.g. vitamin E
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/405Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Definitions

  • the present invention relates to a method of preventing or treating apoptosis using a zinc ionophore.
  • the present invention also relates to a method of protecting cells against the harmful effects of injurious agents, for example, oxidants, TNF ⁇ , neurotoxins, ischemia and radiation.
  • Zinc plays a critical role in cellular biology, and is involved in virtually every important cellular process such as transcription, translation, ion transport, and others (O'Halloran, T.V. (1993) Science 261:715-725; Cousins, R.J. (1994) Annu .Rev. Nutr. 14:449-469; Harrison, ⁇ .L. et al . (1994) Neuropharmacology- 33 :935-952; Berg, J.M. et al . (1996) Science 271:1081-1085).
  • the involvement of cellular zinc in apoptosis has been recognized for close to 20 years (Sunderman, F.W.,Jr. (1995) Ann . Clin. Lab. Sci .
  • Apoptosis is a form of cell death normally activated under physiological conditions, such as involution in tissue remodeling during morphogenesis, and several immunological
  • the apoptotic process is characterized by cell shrinkage, chromatin condensation, and internucleosomal degradation of the cell's DNA (Verh ' aegen et al . (1995) Biochem. Pharmacol. 50(7):1021- 1029) .
  • Zinc-pyrithione (zinc pyridinethione, C 10 H 8 N 2 O 2 S 2 Zn, MW 317.75, commercially available from Sigma) is the active ingredient in the anti-dandruff shampoo Head & Shoulders® (U.S.
  • a method to protect against apoptosis using one or more zinc ionophores In one embodiment of the present invention there is provided a method of treating or preventing apoptosis by administering to a patient in need thereof a pharmaceutically effective amount of a zinc ionophore.
  • a pharmaceutical composition comprising a zinc ionophore and a pharmaceutically acceptable carrier.
  • a method of protecting against the harmful effects of injurious agents selected from the group consisting of oxidants, TNF , neurotoxins, ischemia and radiation by administering to a patient in need of such protection an effective amount of a zinc ionophore.
  • FIGURE 1 shows the effect of zinc pyrithione on rat cardiac apoptosis.
  • FIGURE 2 shows the effects of zinc pyrithione on HSP-70 in heart.
  • FIGURE 3 shows the anti -apoptotic effect of zinc pyrithione on Spl in brain.
  • FIGURE 4 shows the effects of zinc pyrithione on kainic acid induced damage in rat brain areas.
  • FIGURE 5 shows the effects of zinc pyrithione on the severity of kainic acid induced seizures in rats.
  • FIGURE 6 shows the protective effects of zinc pyrithione in PC12 cells subjected to oxidative stress.
  • FIGURE 7 shows the protective effects of zinc pyrithione in PC12 cells.
  • FIGURE 8 shows the anti -apoptotic effect of zinc-pyrithione in irradiated human primary endothelial cells .
  • FIGURE 9 shows the effects of zinc pyrithione on transcription factor binding activity in human primary endothelial cells.
  • FIGURE 10 shows the effects of zinc pyrithione on the TNF -induced transcription factor binding activity in human primary endothelial cells.
  • FIGURE 11 shows the effects of zinc pyrithione on cytosolic Ikappa B protein levels in human primary endothelial cells.
  • FIGURE 12 shows photomicrographs depicting histological evidence of protection by zinc-pyrithione in 4 vessel occlusion stroke model in rats.
  • FIGURE 13 shows the effect of zinc pyrithione on neuronal survival in 4 vessel occlusion stroke model in rats.
  • FIGURE 13A shows the effect of zinc- diethyldithiocarbamate on neuronal survival in 4 vessel occlusion stroke model in rats.
  • FIGURE 14 shows the effect of zinc pyrithione in rats and a 4x reduction in the number of apoptotic nuclei.
  • FIGURE 15 shows middle cerebral artery occlusion caused infarcts in the left hemisphere of mouse brain. Infarcts are visible as white regions in the left hemisphere.
  • FIGURE 16 shows the reduction in infarct area upon treatment with zinc-pyrithione in mice with middle cerebral artery occlusion stroke model.
  • FIGURE 16A shows the reduction in infarct area upon treatment with zinc-diethyldithiocarbamate in mice with middle cerebral artery occlusion stroke model.
  • FIGURE 17 shows the administration of zinc- pyrithione decreased infarct volumes in mouse brain.
  • FIGURE 17A shows the administration of zinc- diethyldithiocarbamate decreased infarct volumes in mouse brain.
  • FIGURE 18 shows the effect of zinc-pyrithione administration on neurological score in mice with middle cerebral artery occlusion stroke model.
  • the present invention is directed to a method of blocking apoptosis.
  • blocking includes treating, preventing, inhibiting, protecting against and reducing the occurrence of apoptosis.
  • treating includes blocking, preventing, inhibiting, protecting against and reducing the occurrence of apoptosis
  • the concentration of zinc ionophore used to block apoptosis ranges from about .005 ⁇ g zinc ionophore per kg of body weight to about 2 mg zinc ionophore per kg of body weight (i.e. about 600pM zinc ionophore to about 6 uM zinc ionophore) .
  • the concentration of zinc ionophore used to block apoptosis ranges from about 1.0 ⁇ g zinc ionophore per kg of body weight to about 800 ⁇ g zinc ionophore per kg of body weight.
  • the concentration of zinc ionophore used to block apoptosis ranges from about 0.2 ⁇ g zinc ionophore per kg of body weight to about 600 ⁇ g zinc ionophore per kg of body weight.
  • the concentration of zinc ionophore used to block apoptosis is about 0.9 mg/kg body weight, or about 0.18mg zinc/kg body weight.
  • any non- toxic compound capable of binding zinc with moderate affinity and having sufficient lipophilic properties to penetrate cell membranes will be capable of effecting the protection demonstrated in the present invention with zinc-pyrithione.
  • zinc- pyrithione was shown to operate at the cell signalling level, as demonstrated by its ability to alter cytosolic PKC- ⁇ content. Further, according to the present invention, the zinc-pyrithione was shown to operate at the transcriptional level, as demonstrated by its ability to alter the nuclear activity of transcription factors NF-kB, AP-1 and Spl . Still further, according to the present invention the zinc-pyrithione was shown to upregulate cytoprotective proteins, for example HSP70. In accordance with the present invention the zinc ionophores protect against neuronal cell loss in stroke patients.
  • zinc pyrithione demonstrates neuroprotective properties, showing protection against cell loss in the selectively vulnerable zone of the CAI region of the hippocampus in a rat model of severe global ischemia. With the mouse model of severe focal ischemia, zinc pyrithione demonstrates neuroprotective properties, significantly decreasing brain infarct volume and neurological deficit.
  • the zinc ionophores are administered in a pharmaceutically effective amount to a patient in need thereof in a pharmaceutical carrier by intravenous, intramuscular, subcutaneous, or intracerebroventricular injection or by oral administration or topical application.
  • one zinc ionophore may be administered, preferably by the intravenous injection route, alone or in conjunction with a second, different zinc ionophore.
  • conjunction with is meant together, substantially simultaneously or sequentially.
  • the zinc ionophores of the present invention are administered acutely, such as, for example, substantially immediately following an injury that results in apoptosis, such as a stroke.
  • the zinc ionophores may therefore be administered for a short course of treatment, such as for about 1 day to about 1 week.
  • the zinc ionophores of the present invention may be administered over a longer period of time to ameliorate chronic apoptotic episodes, such as, for example, for about one week to several months depending upon the condition to be treated.
  • a pharmaceutically effective amount as used herein is meant an amount of zinc ionophore, e.g., zinc- pyrithione, high enough to significantly positively modify the condition to be treated but low enough to avoid serious side effects (at a reasonable benefit/risk ratio) , within the scope of sound medical judgment.
  • a pharmaceutically effective amount of zinc ionophore will vary with the particular goal to be achieved, the age and physical condition of the patient being treated, the severity of the underlying disease, the duration of treatment, the nature of concurrent therapy and the specific zinc ionophore employed. For example, a therapeutically effective amount of a zinc ionophore administered to a child or a neonate will be reduced proportionately in accordance with sound medical judgment. The effective amount of zinc ionophore will thus be the minimum amount which will provide the- desired anti -apoptotic effect.
  • the zinc ionophore e.g. zinc-pyrithione
  • the active ingredients which comprise zinc ionophores may be required to be coated in a material to protect said zinc ionophores from the action of enzymes, acids and other natural conditions which may inactivate said zinc ionophores.
  • they should be coated by, or administered with, a material to prevent inactivation.
  • zinc ionophores may be co- administered with enzyme inhibitors or in liposomes.
  • Enzyme inhibitors include pancreatic trypsin inhibitor, and trasylol .
  • Liposomes include water- in-oil - in-water P40 emulsions as well as conventional and specifically designed liposomes.
  • the zinc ionophores may be administered parenterally or intraperitoneally.
  • Dispersions can also be prepared, for example, in glycerol, liquid polyethylene glycols, and mixtures thereof, and in oils.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the carrier can be a solvent or dispersion medium containing, for example, water, DMSO, ethanol , polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like) , suitable mixtures thereof and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion.
  • a coating such as lecithin
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the zinc ionophore in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized zinc ionophores into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and the freeze-drying technique which yield a powder of the active ingredient plus any additional desired ingredient from previously sterile- filtered solution thereof.
  • the zinc ionophores may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains a zinc ionophore concentration sufficient to treat or block apoptosis in a patient.
  • the tablets, troches, pills, capsules, and the like may contain the following: a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid, and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin may be added or a flavoring agent such as peppermint, oil or wintergreen or cherry flavoring.
  • a binder such as gum tragacanth, acacia, corn starch or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid, and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose or saccharin may be added or a flavoring agent such as pepper
  • a syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor.
  • any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non- toxic in the amounts employed.
  • the zinc ionophore may be incorporated into sustained- release preparations and formulations .
  • pharmaceutically-acceptable carrier as used herein is meant one or more compatible solid or liquid filler diluents or encapsulating substances.
  • compatible as used herein is meant that the components of the composition are capable of being comingled without interacting in a manner which would substantially decrease the pharmaceutical efficacy of the total composition under ordinary use situations.
  • substances which can serve as pharmaceutical carriers are sugars, such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethycellulose, ethylcellulose and cellulose acetates; powdered tragancanth; malt; gelatin; talc; stearic acids; magnesium stearate; calcium sulfate; vegetable oils, such as peanut oils, cotton seed oil, sesame oil, olive oil, corn oil and oil of theobroma; polyols such as propylene glycol, glycerine, sorbitol, manitol, and polyethylene glycol; agar; alginic acids; pyrogen-free water; isotonic saline; and phosphate buffer solution; skim milk powder; as well as other non- toxic compatible substances used in pharmaceutical formulations such as Vitamin C, estrogen and echinacea, for example.
  • Wetting agents and lubricants such as sodium lauryl
  • the patient is administered a therapeutically effective amount of at least one zinc ionophore and a pharmaceutically acceptable carrier in accordance with the present invention.
  • a preferred zinc ionophore is zinc pyrithione.
  • Another preferred zinc ionophore is zinc diethyldithiocarbamate.
  • the zinc ionophores of the present invention are effective against a wide range of injurious agents, for example, but not limited to: oxidants, TNF ⁇ , neurotoxins, or radiation.
  • the zinc ionophores of the present invention are also effective in treating ischemia. Therefore, in a preferred form of treating ischemia the patient is administered a therapeutically effective amount of at least one zinc ionophore and a pharmaceutically acceptable carrier.
  • a preferred zinc ionophore is zinc pyrithione.
  • Another preferred zinc ionophore is zinc diethyldithiocarbamate.
  • compositions suitable for blocking apoptosis which comprise one or more zinc ionophores and a pharmaceutically acceptable carrier.
  • Human umbilical vein endothelial cells were purchased from Clonetics (San Diego, California) and passages 2-4 were used for these studies. Cells were cultured on flame- sterilzed glass coverslips in Endothelial Basal Medium (Clonetics) supplemented with lOng/ml human recombinant epidermal growth factor, 1.0 ug/ml hydrocortisone, 50ug/ml gentamicin, 50ng/ml amphotetericin B, 12ug/ml bovine brain extract and 2%v/v fetal bovine serum (all from Clonetics) , in a humidified chamber at 37°C and 5% C0 2 .
  • Clonetics Endothelial Basal Medium
  • HUVEC proliferating HUVEC were passaged at 80-90% confluency.
  • Cardiac myocytes were isolated from the ventricular septum of adult rabbit hearts, following collagenase digestion, in a manner similar to that described previously (Turan, B. et al., (1997) Am. J. Physiol. 272 :H2095-H2106) .
  • the modification consisted of introducing low concentrations of CaCl 2 during the perfusion with collagenase and the dispersion of the myocytes.
  • Hearts were perfused for about 2 min by gravity under a hydrostatic pressure of 1 m, with a nominally Ca 2+ -free solution containing (in mM) : NaCl, 145; KC1, 5; MgS0 4 , 1.2; Na 2 HP0 4 , 1.8; HEPES, 5; glucose, 10; pH adjusted to 7.4 with NaOH. Forty ml of this perfusate were then supplemented with collagenase (1 mg/ml) and perfusion was continued with recirculation. Within 2-3 min, this treatment resulted in a complete loss of ventricular pressure. The flow rate was then adjusted to 15 ml/min and 50 uM CaCl 2 was added to the collagenase solution.
  • mice Primary cultures of mouse cerebellar granule neurons were obtained from dissociated cerebella of postnatal day 8 or 9 mice according to the following protocol (Cregan et al . , (1999) J. Neurosci . 19:7860- 7869, incorporated herein by reference). Brains were removed and placed into separate dishes containing solution A (124 mM NaCl, 5.37 mM KC1, 1 mM NaH2 P04 , 1.2 mM MgS04 , 14.5 mM D- (1) -glucose, 25 mM HEPES, 3 mg/ml BSA, pH 7.4) in which the cerebella were dissected, meninges removed, and tissue sliced into small pieces.
  • solution A 124 mM NaCl, 5.37 mM KC1, 1 mM NaH2 P04 , 1.2 mM MgS04 , 14.5 mM D- (1) -glucose, 25 mM HEPES, 3 mg/
  • the tissue was briefly centrifuged and transferred to solution A containing 0.25 mg/ml trypsin, then incubated at 37°C for 18 min. After the addition of 0.082 mg/ml trypsin inhibitor (Boehringer Mannheim, Indianapolis, I N) and 0.25 mg/ml DNase I (Boehringer Mannheim), the tissue was incubated at 25°C for 2 min. After a brief centrifugation, the resulting pellet was gently titrated in solution A yielding suspension that was further incubated for 10 min at 25°C in solution A containing 2.7 mM MgS04 and 0.03 mM CaC12. After a final centrifugation the pellet was resuspended in EMEM media (Sigma, St.
  • test compounds with potential zinc- ionophore activity were screened for their ability to transport zinc into selected target cells.
  • the test compounds were first complexed with zinc.
  • the zinc-complexed ionophores holo- ionophores
  • the zinc- free forms of these compounds apo- ionophores
  • purified holo- ionophores were purchased commercially (e.g. zinc-diethyldithiocarbamate, Sigma- Aldrich) . However, in most cases only the apo-ionophores were available commercially.
  • the holo-ionophores were therefore prepared in our laboratory. Since zinc ionophores (e.g. pyrithione, diethyldithiocarbamate, 8- hydroxyquinoline) complex with zinc in a 2:1 molar ratio (ionophore: zinc) , stock solutions (generally 15.7 mM) of holo-ionophores were prepared by combining the apo- ionophore with ZnCl 2 in a 2:1 molar ratio either in water or DMSO, depending on the solubility of the reactants, and incubating at room temperature for 15 min. The holo- ionophores were then stored at -20°C.
  • zinc ionophores e.g. pyrithione, diethyldithiocarbamate, 8- hydroxyquinoline
  • stock solutions generally 15.7 mM
  • test compounds were performed with cultured HUVEC, isolated cardiac myocytes, and cultured cerebellar neurons following an approach described previously (Turan et al . , (1997) Am. J. Physiol. 272 :H2095 -H2106) .
  • the cells were loaded with Fura-2, a zinc and calcium- sensitive indicator, by incubating the cells for 30 min in medium containing 4 uM Fura-2 -am (Molecular Probes) .
  • Glass coverslips bearing HUVEC or cerebellar cells were placed directly in a superfusion chamber on the stage of an epifluorescence inverted microscope (Nikon Diaphot-DM) .
  • the cells were then superfused with superfusion buffer containing a test compound and the fluorescence at 505 nm was recorded in response to excitation at 340 nm and 380 nm.
  • the slope of the fluorescence intensity ratio in response to excitation at 340 and 380 nm was used to determine - ionophore activity.
  • the membrane-permeant heavy metal chelator N,N,N' ,N' , - tetrakis (2 - pyridylmethyl) ethylenediamine (TPEN, 30 uM) was added to the superfusate at the end of the run.
  • Vitamin A (all - trans -retinol) ⁇
  • Vitamin E (alpha- tocopherol) ⁇
  • EXAMPLE 2 In vivo heart model - ischemic injury.
  • the sections were then incubated in 75 ml of a buffer solution containing 200 mmol/L potassium cacodylate, 2 mmol/L CoCl 2 , 0.25 mg/ml bovine serum albumin, 25 mmol/L Tris-HCl, pH 6.6, 10 mmol/L biotin-16- dUTP (Boehringer Mannheim Canada, Laval, Quebec) , and 25 units of terminal transferase (Boehringer) , for 1 h at 37°C in a humidified chamber. The reaction was terminated by washing the sections 3 times (1 min each) with PBS at RT.
  • a buffer solution containing 200 mmol/L potassium cacodylate, 2 mmol/L CoCl 2 , 0.25 mg/ml bovine serum albumin, 25 mmol/L Tris-HCl, pH 6.6, 10 mmol/L biotin-16- dUTP (Boehringer Mannheim Canada, Laval, Quebec) , and 25 units of terminal transfera
  • the sections were then incubated with 1 ml of a staining solution containing 2.5 mg/ml fluorescein isothiocyanate-avidin (avidin-FITC) , 4X saline- sodium citrate buffer, 0.1% Triton X-100, and 5% powdered milk, for 30 min at RT, protected from light.
  • the sections were washed 3 times with PBS, were coverslipped in "anti -fade” solution containing 1 mg/ml p-phenylenediamine, 90% glycerol, in PBS, and histofluorescence was monitored with a Zeiss Axiophot microscope.
  • Positive control samples were prepared by incubating sections with 10 units/ml DNAse I for 20 min at 37°C prior to treatment with terminal transferase.
  • the heart tissue was homogenized on ice for 45 s using a Polytron homogenizer at 10,000 rpm in 8 volumes of 10 mM HEPES (pH 7.9), containing 10 mM KC1, 1.5 mM MgCl 2 , 0.1% Nonidet P-40, 0.5 mM DTT, 0.5 mM PMSF, 0.5 mM spermidine, 0.15 mM spermine, 5 mg/ml aprotinin, 5 mg/ml leupeptin, and 5 mg/ml pepstatin.
  • the homogenate was incubated on ice for 15 minutes and centrifuged at 35,000xg at 4°C for 15 min.
  • PVDF polyvinylidene difluoride
  • HSP70 Since HSP70 has been shown to protect against apoptosis (Wong, H.R., et al . (1996) Am . J. Respir. Cell Mol . Biol . 15:745-751), and to be induced by zinc (Klosterhalfen, B., et al . , (1997) Shock 7:254-262), the data suggest that zinc-pyrithione may exerts its protective effect by upregulating HSP70 synthesis. Zinc-pyrithione also caused a statistically significant decrease in the cytosolic concentration of PKC- ⁇ (data not shown) , using the same method as that described for HSP70, with the exception that instead of using a polyclonal rabbit antibody against HSP70, we used an antibody against PKC- ⁇ . As PKC- ⁇ is a well known mtracellular signalling agent, it was therefore concluded that Zn 2+ is capable of modulating mtracellular signalling.
  • EMSA Electrophoretic Mobility Shift Assay
  • EDTA EDTA
  • 10% glycerol The homogenate was centrifuged at 100,000 xg at 4°C for 1 h, the supernatant was discarded, and the pelleted nuclei were gently resuspended in 40 ml of a lysis buffer containing 20 mM HEPES, pH 7.9, 420 mM NaCl, 1.5 mM MgCl 2 , 0.2 mM EDTA, 25% glycerol, 0.5 mM DTT, 0.5 mM PMSF, 0.5 mM spermidine, 0.15 mM spermine, and 5 mg/ml each of aprotinin, leupeptin and pepstatin.
  • the suspension was incubated on ice for 45 min and centrifuged at 20,000xg at 4°C for 10 min.
  • the supernatant containing nuclear protein was collected and diluted 1:1 with a buffer containing 20 mM HEPES, pH 7.9, 50 mM KC1, 0.2 mM EDTA, 20% glycerol, 0.5 mM DTT, 0.5 mM PMSF, 0.5 mM spermidine, 0.15 mM spermine, and 5 mg/ml each of aprotinin, leupeptin and pepstatin. Protein concentrations were determined using the Bio Rad protein assay.
  • double- stranded consensus oligonucleotides for NF-kB, AP-1 and Spl were radiolabelled with g[ 32 P]ATP (Amersham, Arlington Heights, Illinois) .
  • Five mg of nuclear protein were first incubated for 10 min at room temperature with 5 mg poly-d[I-C] (Boehringer Manheim, Montreal, Quebec) in DNA binding buffer (20 mM HEPES, pH 7.9, 0.2 mM EDTA, 0.2 mM EGTA, 100 mM KC1, 5% glycerol, and 2 mM DTT) .
  • Labeled probe (0.2 ng) was then added and the reaction mix incubated for a an additional 20 min in a final volume of 20 ml.
  • the reaction mixture was subjected to electrophoresis on 5% polyacrylamide gel, and the dried gel was exposed to X-ray film. The intensity of the bands was quantitated with a densitometer and commercially available software (Molecular Analyst, Bio-Rad Laboratories, Hercules, California) .
  • the subunit composition of NF-kB was determined with supershift assays.
  • Antibodies (2 mg) to either p50 or p65 (Santa Cruz Biotechnologies, Inc.
  • Glutamate is the principal excitatory neurotransmitter in the brain, and plays a critical role in the etiology of different major brain pathologies such as cerebral ischemia, neurodegeneration, epilepsy, etc (Coyle, J.T. (1993) Science 262 : 689 - 695) . Compounds which interact with the glutamate receptors are therefore important tools in the investigation of these diseases.
  • Kainate is an excitotoxic glutamate analog that produces excessive neuronal excitation and seizures within hours following its intraperitoneal injection into adult rats. At 2-3 days after treatment, neurodegeneration can be observed in the limbic system in the form of apoptosis
  • the neurotoxic effect of kainate in the rat hippocampal subregions involves a direct effect on presynaptic kainate receptors and an indirect effect on postsynaptic glutamate receptors due to the enhanced release of glutamate (Malva, J.O., et al . , (1998)
  • Zinc pyrithione was injected 15 min after KA administration. Sham operated rats received 1.2% DMSO in the lateral ventricles and isotonic saline mtraperitoneally. ' The volume of substances injected into ventricles was 1.5 uL. Seizure activity in the rats was followed during first 4-6 h after KA administration.
  • Results Brains of sham-operated rats and control animals injected icv with zinc-pyrithione did not show any obvious damage in any brain region. However, administration of KA caused neuronal degeneration and cell loss in a number of brain regions, with injury reaching maximal levels within the first day after KA treatment. Preliminary data suggest that the brain damage is attributable to apoptosis, as detected by the TUNEL stain (not shown) . Twelve rats were subjected to KA treatment alone and 12 were treated with KA followed by zinc-pyrithione. Figure 6 shows the number of rats (out of 12) that displayed detectable signs of injury in each group.
  • Zinc-pyrithione also changed the pattern of KA- induced seizures in the rats.
  • the seizure study was performed with 57 rats in the KA- alone group, and 58 in the KA plus zinc-pyrithione group.
  • the data presented in Figure 5 show the number of rats in which a given seizure severity was the final stage of severity observed.
  • zinc-pyrithione caused a statistically significant 3.2 -fold decrease in the incidence of the most severe and irreversible stage of seizures
  • stage 6 In other words, of the 57 rats treated with KA alone, fully 18 reached the lethal stage 6, as compared with only 5 out of 58 in the KA plus zinc- pyrithione group.
  • the data further show that the final stage of severity tended to be much lower in the zinc- pyrithione group, as illustrated by the 3.2-fold increase in the number of zinc-pyrithione treated rats at level 2 ("wet dog shake” stage). (*, P ⁇ 0.01, Fisher's exact test) .
  • zinc-pyrithione significantly decreased KA- induced cell death in a number of brain regions, and significantly lowered the severity of KA- induced seizures in rats.
  • Oxidative stress is believed to play an important role in the apoptotic neuronal cell death associated with many different neurodegenerative conditions (e.g., Alzheimer's disease, Parkinson's disease, cerebral ischemia, etc.) (Jenner, P. (1994) Lancet 344:796-798).
  • the non-differentiated rat pheochromocytoma PC12 cells are a cell line which differentiates to a neuronal cell type in the presence of Nerve Growth Factor (NGF) , but undergoes apoptotic cell death when deprived of NGF. These cells also undergo apoptotic cell death when exposed to oxidants such as hydrogen peroxide (Satoh, T., et al . , (1997) J. Neurosci . Res . 50:413-420; Maroto, R. et al . , (1997)
  • PC12 cells are therefore a useful cell model with which to analyze the molecular mechanisms of apoptosis induced by oxidative stress and other stimuli in neuronal cells (Kubo, T.,et al . , (1996) Brain Res . 733:175-183, incorporated herein by reference).
  • PC12 cells were seeded on coverslips covered with poly-L- lysine in 24 -well plates, and were grown in RPMI-1640 containing 10% FCS, 5% horse serum, 2 mM glutamine, and 40 mg/kg gentamicin in 5% C0 2 at 37°C.
  • RPMI-1640 medium containing 1% serum (embryonal calf and horse serum, 2:1) and 50 ng/ml NGF was used to induce differentiation.
  • Differentiated PC12 monolayers were washed and were induced to undergo apoptosis in two ways: a) incubation for 4 h with normal growth medium without serum or NGF, and b) incubation for 4 h with normal growth medium (plus serum and NGF) containing 20 ⁇ M hydrogen peroxide (H 2 0 2 ) .
  • the protective effect of zinc- pyrithione against both types of apoptosis was tested by preincubating the cells with this compound for only 5 minutes immediately prior to initiating apoptosis - inducing treatments a or b. Control cells were pretreated with the carrier DMSO alone.
  • Pretreatment with zinc-pyrithione rather than the more relevant post- treatment approach, was used in this model because of practical experimental considerations.
  • the close temporal proximity of the pretreatment to the initiation of the injurious treatment is more representative of a concomitant exposure of the cells to both zinc-pyrithione and the injurious agent, rather than an authentic pretreatment.
  • the cells were fixed with methanol : acetone (1:1) at -20°C and were stained with Hoechst 33258 to> visualize the nuclei.
  • ionizing radiation has been shown to induce the activation of the transcription factor NF-kB in several cell types (Valerie, K. , et al . , (1995) Biochemistry 34:15768-15776), including endothelial cells (Hallahan, D. et al . , (1995) Biochem . Biophys . Res . Commun . 217:784-795).
  • the cytokine TNF ⁇ is also capable of causing apoptosis in endothelial cells either alone
  • the studies performed with this model were designed to examine the ability of zinc- pyrithione to block radiation- induced apoptosis, and to elucidate the effect of zinc-pyrithione at the transcriptional level in response to either ionizing radiation or TNF ⁇ .
  • HUVEC Human umbilical vein endothelial cells
  • Clonetics San Diego, California
  • Cells were cultured on gelatin- coated culture dishes in Endothelial Basal Medium (Clonetics) supplemented with lOng/ml human recombinant epidermal growth factor, 1.0 ug/ml hydrocortisone, 50ug/ml gentamicin, 50ng/ml amphotetericin B, 12ug/ml bovine brain extract and 2%v/v fetal bovine serum, in a humidified chamber at 37°C and 5% C0 2 . To maintain cell populations, proliferating HUVEC were passaged at 80-90% confluency.
  • HUVEC HUVEC were grown to confluency, and then given an additional 24 hours to achieve quiescence prior to experimental treatment. The following treatments were performed:
  • TNF ⁇ The cells were washed twice with 37°C D-PBS and then irradiated in fresh media. Irradiated cells received a dose of 1000 Rads of gamma- irradiation from a 137 Cesium source (Atomic Energy of Canada) . The cells were then incubated for 2 hours (cytosolic and nuclear protein extraction) or 8 hours (Hoechst staining and DNA electrophoresis) .
  • TNF ⁇ The cells were washed twice with 37°C D-PBS and then incubated in media containing the TNF ⁇ (20ng/ml, from a stock of lOug/ml prepared in phosphate buffered saline (PBS) - l%bovine serum albumin. Control cells received fresh media alone. The cells were then incubated for 2 hours (cytosolic and nuclear protein extraction) or 8 hours (Hoechst staining and DNA electrophoresis) .
  • Hoechst Staining Cells were grown on round, gelatin coated 12 mm glass coverslips, and following treatment, were fixed with 0.5 ml of 1% glutaraldehyde in PBS for 10 minutes at room temperature (RT) . The cells were then washed twice with PBS for 5 minutes, and permeabilized with 0.5 ml of 1:1 methanol/acetone for 10 minutes at RT, followed by two five minute PBS washes. The cells were then incubated with Hoechst 33258 (bis-benzimide, 0.05mg/ml in H 2 0) , a fluorescent DNA binding dye, for 30 minutes at room temperature, in the dark. The nuclear morphology of the cells was then visualized under a Zeiss Axiophot fluorescence microscope.
  • the supernatant containing residual cytosolic proteins was discarded and the pelleted nuclei were resuspended in 35ul of Buffer B (20mM HEPES, 420mM NaCl, 1.5mM MgCl 2 , 0.2mM EDTA, 25% glycerol, pH 7.9, 0.5mM DTT, 0.5mM PMSF, and the protease inhibitors spermidine, spermine, aprotinin, leupeptin and pepstatin) for 45 minutes on ice in order to extract the nuclear proteins.
  • Buffer B 20mM HEPES, 420mM NaCl, 1.5mM MgCl 2 , 0.2mM EDTA, 25% glycerol, pH 7.9, 0.5mM DTT, 0.5mM PMSF, and the protease inhibitors spermidine, spermine, aprotinin, leupeptin and pepstatin
  • the protein concentration in the nuclear and cytosolic extracts was determined using the Bradford Assay (Biorad) using bovine serum albumin as the standard.
  • Electrophoretic Mobility Shift Assay Equal amounts of nuclear protein (5ug) were incubated with poly dl-dC (5ug from a stock of 2.5ug/ul in TE buffer) for 10 minutes at RT. This reaction mixture was then incubated with 0.2ng of 5' end- 32 phosphorus - labelled double stranded oligonucleotide probe for 20 minutes at RT to allow the binding of nuclear proteins with the labeled probe.
  • Loading buffer (5ul of a mixture containing 20mM HEPES, lOOmM KC1, 60% glycerol, 0.5mM EDTA, 0.5mM EGTA and 0.125% bromophenol blue) was added to the reaction mixture prior to the electrophoresis on a 5% native polyacrylamide gel.
  • the gels were run in Tris-Glycine solution for 1.5 hours at 200V and were then dried between filter paper and cellophane for 1.5 hours at 80°C under vacuum. The dried gels were exposed to X-ray film (Cronex) for up to 2 days at -80°C.
  • reaction mixture was incubated with a 125- fold excess of unlabeled probe for 20 minutes at RT prior to the addition of the labeled probe.
  • reaction mixture was incubated with 2mg of rabbit polyclonal anti-NFkB p50 or p65 antibody (Santa Cruz Biotechnology) for 20 minutes at RT immediately subsequent to the addition of the labeled probe. The bound antibody retards the mobility of the protein-DNA complex, resulting in a shifted band.
  • the consensus oligonucleotides for the transcription factors NFkB (5' - ACT TGA GGG GAC TTT CCC AGG C - 3 ' ) , AP - 1 ( 5 ' - CGC TTG ATG AGT CAG CCG GAA-3') and Spl (5' -ATT CGA TCG GGG CGG GGC GAG C-3') (Promega) and were labeled as suggested by Promega with minor modifications.
  • oligonucleotides (20ng) , T4 Polynucleotide kinase and [g32P]ATP (60uCi) were mixed in kinase buffer (50mM Tris- HCl, pH 7.6, lOmM MgCl 2 , 5% glycerol and 5mM DTT) and incubated at 37°C for 1 hour. Labeled oligonucleotides were removed by centrifugation through a G-25 Sephadex Column at 8500rpm for 20 minutes. The labeled oligonucleotides were then diluted such that 2ul of the probe mixture contained approximately 50000 - lOOOOOcpm.
  • the blots were then washed with TBS-T and incubated for 1.5 hours in primary antibody (anti-IkBa, Santa Cruz Biotechnology) diluted 1:1000 in 2% skimmed milk in TBS-T and sodium azide at RT with constant shaking. The blots were then washed with TBS-T and incubated for 30 minutes in horseradish peroxidase labeled goat anti -rabbit IgG diluted 1:10000 in 2% skimmed milk in TBS-T at RT with constant shaking. Following treatment with the secondary antibody, the blots were extensively washed with TBS-T and incubated for 1 minute with chemiluminescent substrate. The blots were then exposed to X-ray film for 1-5 minutes.
  • primary antibody anti-IkBa, Santa Cruz Biotechnology
  • Results The data show protection by zinc-pyrithione against endothelial apoptosis, and also show that this protective effect is associated with transcriptional modulation.
  • the data also show that zinc is required for this effect since the sodium salt of pyrithione was not effective in preventing apoptosis.
  • DMSO alone was also ineffective. No apoptosis was caused in control cultures by zinc-pyrithione, sodium-pyrithione, or DMSO alone ( Figure 8).
  • (*, P ⁇ 0.05 vs. Control; °, P ⁇ 0.05 vs. irradiated cells, n 4).
  • EMSA tests showed that irradiation- induced apoptosis is associated with a significant increase in nuclear NF-kB content, and that zinc-pyrithione, but not sodium-pyrithione or DMSO alone, blocked this increase (Figure 9) .
  • Zinc-pyrithione had a very similar effect in TNF ⁇ -treated HUVEC ( Figure 10), and was particularly potent at blocking the TNF -induced increase in NF-kB content.
  • Stroke is an extremely variable clinical condition which reflects the variability of the underlying disease process.
  • the vascular occlusion can occur at many different sites in the brain and the cause of the occlusion, the severity of the problem, and the degree of reversibility can all contribute to the variability of outcome.
  • most of these variables can be controlled or eliminated, enabling a meaningful interpretation of the results.
  • stroke models are grouped into those producing either global or focal ischemia
  • ZP zinc pyrithione
  • 4VO a global ischemia -reperfusion model of 4 vessel occlusion in rats
  • MCAO middle cerebral artery occlusion in mice
  • the 4VO approach sealed off the two carotid and two vertebral arteries which carry all the blood to the brain. This approach permitted severe forebrain ischemia to be produced in awake and freely moving rats, and produced reproducible neuropathology.
  • the 4VO is a two-stage operative procedure (Pulsinelli, et al . , (1979) Stroke, 10: 267-272, incorporated herein by reference) .
  • mice the 4VO is a favorite stroke model because it results in highly reproducible damage in the CAI region of the hippocampus, as well as in some other brain regions.
  • MCAO in mice is one of the most clinically relevant stroke models. It shuts off blood flow to only a portion of the brain, producing a focal injury which closely resembles the clinical situation with stroke patients. This procedure was performed with an intraluminal thread. A nylon suture was introduced into the external carotid artery and was gently advanced into the internal carotid artery. The diameter of the suture was such that it lodged in the anterior cerebral artery, occluding the medial cerebral artery at its origin.
  • Brain damage in this model was observed as early as several hours after the ischemic episode, with optimal injury occurring at 24 h.
  • the injury occupied a large part of the hemisphere including the cerebral cortex and subcortical structures, and its severity depended on the duration of ischemia and the strain of mice used.
  • the 4VO procedure was performed on male Wistar rats weighing 240-300 g.
  • the rats were housed in groups of 5 in plastic cages with free access to food and water.
  • the MCAO protocol was performed on male C57BL/6 mice weighing 20-28 g which were housed in groups of 10 in plastic cages with free access to food and water. All experiments were performed in accordance with the National Institutes of Health Guidelines for the Care and Use of Laboratory Animals.
  • Rats under chloral hydrate anesthesia (325 mg/kg) were positioned in a stereotaxic frame.
  • the vertebral arteries were exposed and permanently occluded by electrocautery at the first cervical vertebra.
  • Snares surgical silk strings
  • the animals were then allowed to recover for 24 hours with free access to water.
  • the rats were lightly anesthetized with ether, were secured to surgical boards, ventral side up, and their common carotid arteries were exposed.
  • Forebrain ischemia was initiated by tightening the snares around the carotid arteries for 10 min.
  • the body temperature of the rats was carefully maintained at ⁇ 37-37.5°C, both before and during the ischemic insult, using a feedback- controlled heating pad and a rectal thermistor (Homeothermic Blanket System, Harvard Apparatus LTD, England) .
  • the initial (1 st min of ischemia) and final (10 th minute of ischemia) temperature did not differ among all groups of rats studied.
  • the temperature was maintained in similar fashion at 37°C for at least 4 h. Only rats that showed signs of severe neurological injury, such as a loss of the righting reflex, pupil dilation, etc., were included in this study.
  • the vertebral and carotid arteries were exposed, but were not occluded.
  • Evaluations of neurological deficit were performed at 24 and 96 h after ischemia, and were based on a scoring system which recorded activity level, motility, pain reflex, grabbing reflex, and the ability to see and hear (Miljkovic, L.M., et al . , (1997) Ann. Emerg. Med. 29, 758-765, incorporated herein by reference).
  • mice C57BL/6 were anesthetized with an intraperitoneal injection of chloral hydrate (350 mg/kg) and xylasine (4 mg/kg) .
  • Focal cerebral ischemia was produced by occlusion of the MCA using the intraluminal filament technique (Longa, Z.E., et al . , (1989) Stroke 20 : 84 -91, incorporated herein by reference).
  • a 8.0 nylon microfilament coated with a silicon resin (Xantopren) - hardener mixture Hara, H., et al . , (1996) J. Cereb. Blood Flow Metab.
  • Stroke 17:472-476 incorporated herein by reference: 0, no observable neurological deficit (normal); 1, failure to extend the right forepaw (mild); 2, circling to the contralateral side (moderate); 3, falling to the right (severe); 4, inability to walk spontaneously (most severe) .
  • ZP zinc pyrithione
  • ZnDDC zinc- diethyldithiocarbamate
  • control and experimental animals were deeply anaesthetized with sodium thiopental (60 mg/kg, mtraperitoneally) and were perfused transcardially with 250 ml of AFA fixative (96% alcohol, 39 % formalin, glacial acetic acid , 7:2:1). After the AFA perfusion the heads were collected intact and were kept at 4°C for 4-5 h. The brains were then removed and immersed in the same fixative for 1 h, and were then stored in 70% alcohol. Each forebrain was cut into three frontal blocks and imbedded in paraffin. Ten ⁇ m thick sections were cut from a region 3.0-4.0 mm posterior to the bregma. The sections were stained with cresyl violet (Nissl) .
  • ISEL In Situ End Labeling
  • MCAO Mice were killed 24 h after reperfusion with an overdose of sodium thiopental (60 mg/kg, mtraperitoneally) , and the brain was rapidly removed and sectioned coronally into five 1.7 mm slices. The slices were then placed in 2% (wt/vol) 2,3,5- triphenyltetrazolium chloride solution (TTC) in PBS (pH 7.4) for 20 min at 37°C. This procedure, which tests mitochondrial activity, stains viable tissue a bright red, while the infarcted regions remain white. Following TTC staining the sections were fixed in 10% formalin overnight. The area of infarct in each section was determined using an image-analysis system.
  • TTC 2,3,5- triphenyltetrazolium chloride solution
  • the infarct volume was subsequently calculated by summing the infarct areas in the sequential 1.7 mm- thick sections with correction for edema.
  • the person measuring infarct volumes was blinded as to the identity of the experimental groups.
  • ISEL staining the brains were removed, were frozen rapidly, and were sectioned with a cryostat into 20 urn sections from the anterior side to the posterior side at 500 urn intervals. ISEL was performed as described above. Adjacent sections were stained with cresyl violet (Nissl) .
  • the ApopTag® Peroxidase In situ Apoptosis detection Kit (Intergen) was also used to detect apoptosis in mouse brain sections .
  • the neurological deficit data in 4VO rats are presented in Table 2.
  • a method of scoring in which increasing neurological scores are indicative of decreasing neurological function compared to a perfect score of 0 for the shams was employed.
  • a large number of the 4VO rats did not show any neurological deficit (score 0) , despite the fact that they sustained an almost complete loss of cells in the CAl region. Therefore, the administration of ZP did not influence the neurological deficit in the 4VO rats despite clear evidence of histological protection.
  • the neurological deficit data for this ZP administration schedule are presented in Table 2.
  • the neurological score in "3, 4, 6 h schedule" rats did not differ from that in the other regimen (10 min, 1 h, 2 h after ischemic episode) .
  • These data therefore indicated that delaying the first administration of ZP by 3 hours did not significantly change its neuroprotective effect at the dose of 3x6 ⁇ g/kg, indicating a possible wide therapeutic window for ZP in the 4VO stroke model.
  • MCAO MCAO for 1 h produced significant infarcts in the left hemisphere of mouse brain (Figure 15) .
  • Three doses of ZP were used (3x1.2 ⁇ g/kg, 3x6 ⁇ g/kg, and 3x30 ⁇ g/kg) and infarct areas, infarct volumes and neurological scores were measured.
  • ZP at all three doses significantly decreased the infarct area at a distance of 3.4-5.8 mm from the frontal pole ( Figure 16) .
  • Table 3 presents data on the absolute and relative (% of the contralateral hemisphere) infarct volumes in the mouse MCAO model in response to zinc pyrithione treatment.
  • the calculated infarct volumes in the ischemic control mice were similar to those reported previously for this strain of mouse (Hara, H. , et al . (1996) J. Cereb. Blood Flow Metab. 16:605-611; MacManus, J.P., et al., (1999) NeuroReport 10:2711-2714; Nagayama, M., et al., (1999) J Cereb. Blood Flow Metab. 11:1213- 1219; Nogawa, S., et al . , (1998) Proc . Natl .
  • the pericardium will be opened and fixed to the border of the sternum.
  • a left atrial pressure (LAP) catheter will be introduced.
  • Coronary occlusion by a snare will be applied by tunneling the left anterior descending (LAD) coronary artery with a monofil suture between the proximal and medial third behind the first diagonal branch.
  • LAD left anterior descending coronary artery
  • the vena cordis magna will be cannulated for blood analysis with a small catheter.
  • a myocardial P0 2 probe will then be implanted into the expected center of the area at risk.
  • a temperature probe will be positioned next to the P0 2 sensor. Baseline values will be acquired during a 1-hour preoperative period.
  • Coronary occlusion will be achieved by tightening the snare around the LAD for 60 min. The snare will then be loosened to initiate reperfusion.
  • Two models will be tested: I. An acute non- survival model with a total of 6 h reperfusion, and II. A recovery model with 7 days of reperfusion. Each of the two models will consist of 5 groups: Sham operated (Sham), Ischemia/reperfusion alone (I/R) , I/R plus vehicle alone (I/R + DMSO) , I/R plus zinc pyrithione (I/R + ZP) , and I/R plus Zinc-diethyldithiocarbamate (I/R + ZD) .
  • the zinc ionophores will be infused into the LAD in three boluses of 5ml saline, at 0, 1, and 2h after the initiation of reperfusion to give a cumulative dose of 1 ⁇ g/kg body weight.
  • Vehicle will be administered in a similar and blinded fasion.
  • Hemodynamic and P0 2 measurements and blood samples will be obtained at 5 , 10, 20, 30, and 60 minutes after coronary occlusion; and after 5, 10, 20, 30, 60, 90, and 120 minutes of coronary reperfusion.
  • Global and regional contractility will be recorded by regional 2Dultrasound before, after 15 and 60 minutes of coronary occlusion, and after 15, 60, and 120 minutes of reperfusion.
  • the pigs will be killed, and their hearts will be recovered for further analysis .
  • ECG, right atrial, pulmonary artery, and arterial pressure and LAP will be recorded on a Siemens Sirecust 404-1 at different time points.
  • Cardiac output will be determined by thermodilution (5 mL NaCl 0.9%, room temperature) and by continuous measurement with a Baxter Vigilance monitor.
  • the Licox catheter probe measurement system will be used in which the flexible
  • Licox catheter P0 2 microprobe is in direct contact with the myocardium.
  • One milliliter of heparinized venous and arterial blood will be drawn with polypropylene syringes from the femoral artery and the vena cordis magna. Lactate and arteriovenous 0 2 differences will be determined before, after 60 minutes of coronary occlusion, and after 10 and 120 minutes of reperfusion.
  • Blood gas analysis will be performed with the Radiometer Copenhagen Arterial Bloodgas Laboratory 3.
  • samples will be centrifuged at 2000g (10 minutes at 4°C) , plasma will be decanted, and lactate will be measured with the Lactate Analyzer model 23L from Yellow Springs Instrument Co Inc.
  • Venous blood samples (3 ml) will be collected in polypropylene tubes containing citrate and will be centrifuged at 2000g for 15 minutes at 4°C. Plasma creatine kinase activity will be determined and expressed as international units per milliliter. Troponin-T will be measured.
  • Unstained myocardium will be defined as the area at risk. After cardioplegia with 20 mL potassium chloride IV (20%) , the heart will be excised. The right ventricle, the large vessels, and fat tissue will be removed. The left ventricle will then be sliced perpendicular to the axis of the left side of the heart from the apex to the AV groove in 4 -mm slices.
  • the unstained part of the left ventricular myocardium will be separated from the Evan's blue-stained portion and immersed in a 0.09-mol/L sodium phosphate buffer, pH 7.4, containing 1% triphenyltetrazolium chloride (TTC, Sigma) and 8% dextran (molecular weight, 77.800) for 20 minutes at 37°C.
  • TTC triphenyltetrazolium chloride
  • the TTC dye will form a dark-red formazan complex in the presence of viable myocardial cells that contain active dehydrogenases and cofactors. Dead cells will remain unstained.
  • the ischemic but non-necrotic, red- stained tissue will be separated from the unstained, infarcted tissue.
  • the three tissue sections—nonischemic (area not at risk) , ischemic non-necrotic, and ischemic necrotic tissue will be weighed.
  • the arterial pressure-rate product (mean arterial pressure times heart rate) will be taken as a global parameter of myocardial contractility.
  • TUNEL staining (Fliss et al . (1996) supra) , as described above, will be used to identify the percent of apoptotic myocytes in the myocardial tissue.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Emergency Medicine (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The present invention provides methods and compositions comprising one or more zinc ionophores for blocking apoptosis in patients in need thereof. Concentrations of zinc-pyrithione and diethyl-dithiocarbamate in the picomolar to nanomolar range have a strong protective effect against apoptosis.

Description

ZINC IONOPHORES AS ANTI-APOPTOTIC AGENTS
The present invention relates to a method of preventing or treating apoptosis using a zinc ionophore. The present invention also relates to a method of protecting cells against the harmful effects of injurious agents, for example, oxidants, TNFα, neurotoxins, ischemia and radiation.
Zinc plays a critical role in cellular biology, and is involved in virtually every important cellular process such as transcription, translation, ion transport, and others (O'Halloran, T.V. (1993) Science 261:715-725; Cousins, R.J. (1994) Annu .Rev. Nutr. 14:449-469; Harrison, Ν.L. et al . (1994) Neuropharmacology- 33 :935-952; Berg, J.M. et al . (1996) Science 271:1081-1085). The involvement of cellular zinc in apoptosis has been recognized for close to 20 years (Sunderman, F.W.,Jr. (1995) Ann . Clin. Lab. Sci . 25:134-142; Fraker, P.J. et al. (1997) Proc. Soc. Exp . Biol .Med. 215:229-236.). However, the full nature of this involvement is not fully understood. Apoptosis is a form of cell death normally activated under physiological conditions, such as involution in tissue remodeling during morphogenesis, and several immunological
processes. The apoptotic process is characterized by cell shrinkage, chromatin condensation, and internucleosomal degradation of the cell's DNA (Verh'aegen et al . (1995) Biochem. Pharmacol. 50(7):1021- 1029) .
Numerous in vitro studies have been done recently in an attempt to elucidate the role of mtracellular zinc. Although some studies have suggested that zinc may actually induce apoptosis (Xu, J. et al . (1996) Am.J.Physiol. 270:G60-G70; Kim, Y.H. et al . ,
(1999) Neuroscience 89:175-182), most have concluded that increasing the mtracellular concentrations of zinc blocks apoptosis (Sunderman, F.W.,Jr. (1995) Ann.Clin.Lab.Sci. 25:134-142; Adebodun, F. et al . (1995) J.Cell.Physiol. 163:80-86; Zalewski, P.D., et al . (1993) Biochem. J. 296:403-408), and that decreasing the zinc concentration promotes apoptosis (Jiang, S., et al. (1995) Lab. Invest. 73:111-117; Treves, S., et al . (1994) Exp.Cell Res. 211:339-343; Ahn, Y.H., et al . (1998) Exp.Neurol. 154:47-56). The manner in which increased mtracellular zinc affords protection against apoptosis is not clear. (Truong-Tran, A.Q. et al . , (2000) J. Nutr. 130:1459S-1466S) One theory proposes that zinc inactivates the mtracellular endonuclease (s) responsible for apoptotic DNA fragmentation (Shiokawa, D., et al .
(1994) Eur.J. Biochem. 226:23-30; Yao, M. et al . , (1996) J. Mol. Cell. Cardiol. 28:95-101). Other recent studies have suggested that zinc can inhibit caspases (Jiang, S., et al. (1997) Cell Death Differ. 4:39-50; Perry, D.K., et al. (1997) J.Biol.Chem. 272:18530-18533; Maret, W. , et al. (1999) Proc. Natl.Acad. Sci. USA 96:1936-1940) , or block the activation of caspases (Aiuchi, T., et al . (1998) J. Biochem . 124:300-303). However, in view of the large number of mtracellular roles played by zinc, it seems likely that its anti- apoptotic mechanisms may be more complex, possibly involving gene expression and cellular signalling pathways. In fact, recent studies support a role for zinc transients in mtracellular signalling and gene expression (O'Halloran, T.V. (1993) Science 261:715-725; Berg, J.M. , et al . , (1996) Science 271:1081-1085) . In contrast to the large number of in vi tro studies, very few studies have attempted to examine the protective effects of zinc in vivo . It is important to note that most of the studies that have explored this possibility have focused on the pretreatment of tissues with zinc prior to injury. Using this approach, a number of studies have demonstrated that pretreatment of animals with zinc at least 24 hours prior to injury provided some measure of protection against apoptosis (Thomas, D.J. et al., (1991) Toxicology 68:327-337; Matsushita, K. , et al., (1996) Brain Res . 743:362-365; Klosterhalfen, B., et al . , (1997) Shock 7:254-262), presumably as a result of the well established ability of zinc to boost the immune system (Cunningham-Rundles, S., et al . , (1990) Ann . N. Y. Acad. Sci . 587:113-122). Also, one study showed that several days of zinc dietary supplementation concomitant with i.p. injection of carbon tetrachloride protected against liver apoptosis (Cabre, M. et al . (1999) J. Hepa tol . 31:228-234). However, no studies have demonstrated the efficacy of zinc when administered acutely and post- injury, a much more clinically relevant setting. Zinc-pyrithione (zinc pyridinethione, C10H8N2O2S2Zn, MW 317.75, commercially available from Sigma) is the active ingredient in the anti-dandruff shampoo Head & Shoulders® (U.S. patents 3,236,733, and 3,281,366, both 1966), as well as a number of other topical skin treatment formulations. It is a fungicide and bactericide at high concentrations. It is highly lipophilic and therefore penetrates membranes easily. This permits zinc pyrithione to transport zinc across cell membranes, thereby conferring on this compound (i.e. zinc pyrithione) the properties of a zinc ionophore. The anti -apoptotic effect of zinc pyrithione was first observed in vi tro by Zalewski and coworkers , who showed that micromolar concentrations of this compound protected lymphocytic leukemia cells against colchicine- induced apoptosis (Giannakis, C, et al . (1991)
Biochem . Biophys . Res . Commun . 181:915-920). The rationale for the use of this zinc ionophore was to facilitate the transport of Zn+ into the target cells. This is necessitated by the fact that all eukaryotic cells strictly regulate the membrane transport of Zn2+, making it very difficult to modulate the mtracellular concentration and distribution of Zn2+. Zalewski 's group has since published a number of other studies, all of them in vi tro, confirming the ability of micromolar concentrations of zinc-pyrithione to rapidly transport Zn2+ into cells and to thereby prevent apoptosis (Zalewski, P.D., et al . (1994) supra; Zalewski, P.D., et al. (1993) Biochem. J. 296:403-408). One confirmatory study, also in vi tro, has been published from another laboratory (Tempel, K.-H. et al . , (1993) Arch . Toxicol . 67:318-324). In addition to zinc-pyrithione, another group of zinc ionophores, the dithiocarbamates, has been shown to affect apoptosis in vi tro. (Orrenius, S. et al . (1996) Biochem . Soc . Trans . 24:1032-1038; Stefan, C. et al. (1997) Chem . Res . Toxicol . 10:636-643; Erl, W. et al., (2000) Am . J. Physiol . 278 :C1116 -C1125) . However, no attempts have been made to examine in vivo the protective effects of zinc-pyrithione, zinc-bound dithiocarbamates or any other known zinc ionophore at nanomolar or picomolar concentrations.
The ability of zinc-pyrithione and zinc- diethyldithiocarbamates to protect against apoptosis in three models of in vivo injury, as well as two in vi tro models is presented. In each case a pronounced anti- apoptotic effect was achieved.
Thus, according to the present invention there is provided a method to protect against apoptosis using one or more zinc ionophores. In one embodiment of the present invention there is provided a method of treating or preventing apoptosis by administering to a patient in need thereof a pharmaceutically effective amount of a zinc ionophore.
In another embodiment of the present invention there is provided a pharmaceutical composition comprising a zinc ionophore and a pharmaceutically acceptable carrier.
In a further embodiment of the present invention there is provided a method of protecting against the harmful effects of injurious agents selected from the group consisting of oxidants, TNF , neurotoxins, ischemia and radiation by administering to a patient in need of such protection an effective amount of a zinc ionophore.
These and other features of the invention will become more apparent from the following description in which reference is made to the appended drawings wherein: FIGURE 1 shows the effect of zinc pyrithione on rat cardiac apoptosis. FIGURE 2 shows the effects of zinc pyrithione on HSP-70 in heart.
FIGURE 3 shows the anti -apoptotic effect of zinc pyrithione on Spl in brain.
FIGURE 4 shows the effects of zinc pyrithione on kainic acid induced damage in rat brain areas.
FIGURE 5 shows the effects of zinc pyrithione on the severity of kainic acid induced seizures in rats.
FIGURE 6 shows the protective effects of zinc pyrithione in PC12 cells subjected to oxidative stress. FIGURE 7 shows the protective effects of zinc pyrithione in PC12 cells.
FIGURE 8 shows the anti -apoptotic effect of zinc-pyrithione in irradiated human primary endothelial cells . FIGURE 9 shows the effects of zinc pyrithione on transcription factor binding activity in human primary endothelial cells.
FIGURE 10 shows the effects of zinc pyrithione on the TNF -induced transcription factor binding activity in human primary endothelial cells. FIGURE 11 shows the effects of zinc pyrithione on cytosolic Ikappa B protein levels in human primary endothelial cells.
FIGURE 12 shows photomicrographs depicting histological evidence of protection by zinc-pyrithione in 4 vessel occlusion stroke model in rats.
FIGURE 13 shows the effect of zinc pyrithione on neuronal survival in 4 vessel occlusion stroke model in rats. FIGURE 13A shows the effect of zinc- diethyldithiocarbamate on neuronal survival in 4 vessel occlusion stroke model in rats.
FIGURE 14 shows the effect of zinc pyrithione in rats and a 4x reduction in the number of apoptotic nuclei.
FIGURE 15 shows middle cerebral artery occlusion caused infarcts in the left hemisphere of mouse brain. Infarcts are visible as white regions in the left hemisphere. FIGURE 16 shows the reduction in infarct area upon treatment with zinc-pyrithione in mice with middle cerebral artery occlusion stroke model.
FIGURE 16A shows the reduction in infarct area upon treatment with zinc-diethyldithiocarbamate in mice with middle cerebral artery occlusion stroke model.
FIGURE 17 shows the administration of zinc- pyrithione decreased infarct volumes in mouse brain.
FIGURE 17A shows the administration of zinc- diethyldithiocarbamate decreased infarct volumes in mouse brain. FIGURE 18 shows the effect of zinc-pyrithione administration on neurological score in mice with middle cerebral artery occlusion stroke model.
The present invention is directed to a method of blocking apoptosis. In accordance with the present invention "blocking" includes treating, preventing, inhibiting, protecting against and reducing the occurrence of apoptosis. Likewise and in accordance with the present invention "treating" includes blocking, preventing, inhibiting, protecting against and reducing the occurrence of apoptosis
Previous studies have shown that treatment of injured cells with zinc can block apoptosis. In these ' studies the effectiveness of zinc ionophore was demonstrated using high concentrations, ranging from 3μM to 5mM. It was found, according to the present invention that such concentrations are not suitable for use in any in vivo method. Thus, according to the present invention, small concentrations of a zinc ionophore in the nanomolar and picomolar range, such as from about 10 pM to about 1μM, to block apoptosis.
Thus, according to the present invention the concentration of zinc ionophore used to block apoptosis ranges from about .005 μg zinc ionophore per kg of body weight to about 2 mg zinc ionophore per kg of body weight (i.e. about 600pM zinc ionophore to about 6 uM zinc ionophore) . In a further embodiment of the present invention the concentration of zinc ionophore used to block apoptosis ranges from about 1.0 μg zinc ionophore per kg of body weight to about 800 μg zinc ionophore per kg of body weight. Preferably the concentration of zinc ionophore used to block apoptosis ranges from about 0.2μg zinc ionophore per kg of body weight to about 600μg zinc ionophore per kg of body weight. In a further embodiment of the present invention the concentration of zinc ionophore used to block apoptosis is about 0.9 mg/kg body weight, or about 0.18mg zinc/kg body weight.
According to the present invention, any non- toxic compound capable of binding zinc with moderate affinity and having sufficient lipophilic properties to penetrate cell membranes will be capable of effecting the protection demonstrated in the present invention with zinc-pyrithione. The following are examples of compounds which have been shown in accordance with the present invention to possess zinc- ionophore properties: zinc pyrithione, the heterocyclic amines including, for example, 5 , 7 -Diiodo- 8-hydroxyquinoline, and 8- Hydroxyquinoline; the dithiocarbamates including, for example, pyrrolidine dithiocarbamate and diethyldithiocarbamate, disulfiram and dimethyldithiocarbamate; and Vitamins including, but not limited to, Vitamin E and Vitamin A.
According to the present invention zinc- pyrithione was shown to operate at the cell signalling level, as demonstrated by its ability to alter cytosolic PKC-α content. Further, according to the present invention, the zinc-pyrithione was shown to operate at the transcriptional level, as demonstrated by its ability to alter the nuclear activity of transcription factors NF-kB, AP-1 and Spl . Still further, according to the present invention the zinc-pyrithione was shown to upregulate cytoprotective proteins, for example HSP70. In accordance with the present invention the zinc ionophores protect against neuronal cell loss in stroke patients. For example, zinc pyrithione demonstrates neuroprotective properties, showing protection against cell loss in the selectively vulnerable zone of the CAI region of the hippocampus in a rat model of severe global ischemia. With the mouse model of severe focal ischemia, zinc pyrithione demonstrates neuroprotective properties, significantly decreasing brain infarct volume and neurological deficit.
In use, the zinc ionophores, according to the present invention are administered in a pharmaceutically effective amount to a patient in need thereof in a pharmaceutical carrier by intravenous, intramuscular, subcutaneous, or intracerebroventricular injection or by oral administration or topical application. In accordance with the present invention one zinc ionophore may be administered, preferably by the intravenous injection route, alone or in conjunction with a second, different zinc ionophore. By "in conjunction with" is meant together, substantially simultaneously or sequentially. In one embodiment, the zinc ionophores of the present invention are administered acutely, such as, for example, substantially immediately following an injury that results in apoptosis, such as a stroke. The zinc ionophores may therefore be administered for a short course of treatment, such as for about 1 day to about 1 week. In another embodiment, the zinc ionophores of the present invention may be administered over a longer period of time to ameliorate chronic apoptotic episodes, such as, for example, for about one week to several months depending upon the condition to be treated.
By "pharmaceutically effective amount" as used herein is meant an amount of zinc ionophore, e.g., zinc- pyrithione, high enough to significantly positively modify the condition to be treated but low enough to avoid serious side effects (at a reasonable benefit/risk ratio) , within the scope of sound medical judgment. A pharmaceutically effective amount of zinc ionophore will vary with the particular goal to be achieved, the age and physical condition of the patient being treated, the severity of the underlying disease, the duration of treatment, the nature of concurrent therapy and the specific zinc ionophore employed. For example, a therapeutically effective amount of a zinc ionophore administered to a child or a neonate will be reduced proportionately in accordance with sound medical judgment. The effective amount of zinc ionophore will thus be the minimum amount which will provide the- desired anti -apoptotic effect.
A decided practical advantage is that the zinc ionophore, e.g. zinc-pyrithione, may be administered in a convenient manner such as by the, intravenous, intramuscular, subcutaneous, oral or intra- cerebroventricular injection routes or by topical application. Depending on the route of administration, the active ingredients which comprise zinc ionophores may be required to be coated in a material to protect said zinc ionophores from the action of enzymes, acids and other natural conditions which may inactivate said zinc ionophores. In order to administer zinc ionophores by other than parenteral administration, they should be coated by, or administered with, a material to prevent inactivation. For example, zinc ionophores may be co- administered with enzyme inhibitors or in liposomes. Enzyme inhibitors include pancreatic trypsin inhibitor, and trasylol . Liposomes include water- in-oil - in-water P40 emulsions as well as conventional and specifically designed liposomes.
The zinc ionophores may be administered parenterally or intraperitoneally. Dispersions can also be prepared, for example, in glycerol, liquid polyethylene glycols, and mixtures thereof, and in oils.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage. The carrier can be a solvent or dispersion medium containing, for example, water, DMSO, ethanol , polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like) , suitable mixtures thereof and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion. In many cases it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the zinc ionophore in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized zinc ionophores into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and the freeze-drying technique which yield a powder of the active ingredient plus any additional desired ingredient from previously sterile- filtered solution thereof.
For oral therapeutic administration, the zinc ionophores may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains a zinc ionophore concentration sufficient to treat or block apoptosis in a patient.
The tablets, troches, pills, capsules, and the like, may contain the following: a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid, and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin may be added or a flavoring agent such as peppermint, oil or wintergreen or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills or capsules or zinc ionophore in suspension may be coated with shellac, sugar or both.
A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non- toxic in the amounts employed. In addition, the zinc ionophore may be incorporated into sustained- release preparations and formulations .
By "pharmaceutically-acceptable carrier" as used herein is meant one or more compatible solid or liquid filler diluents or encapsulating substances. By "compatible" as used herein is meant that the components of the composition are capable of being comingled without interacting in a manner which would substantially decrease the pharmaceutical efficacy of the total composition under ordinary use situations. Some examples of substances which can serve as pharmaceutical carriers are sugars, such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethycellulose, ethylcellulose and cellulose acetates; powdered tragancanth; malt; gelatin; talc; stearic acids; magnesium stearate; calcium sulfate; vegetable oils, such as peanut oils, cotton seed oil, sesame oil, olive oil, corn oil and oil of theobroma; polyols such as propylene glycol, glycerine, sorbitol, manitol, and polyethylene glycol; agar; alginic acids; pyrogen-free water; isotonic saline; and phosphate buffer solution; skim milk powder; as well as other non- toxic compatible substances used in pharmaceutical formulations such as Vitamin C, estrogen and echinacea, for example. Wetting agents and lubricants such as sodium lauryl sulfate, as well as coloring agents, flavoring agents, lubricants, excipients, tableting agents, stabilizers, anti-oxidants and preservatives, can also be present.
Accordingly, in a preferred form of blocking apoptosis the patient is administered a therapeutically effective amount of at least one zinc ionophore and a pharmaceutically acceptable carrier in accordance with the present invention. A preferred zinc ionophore is zinc pyrithione. Another preferred zinc ionophore is zinc diethyldithiocarbamate.
The zinc ionophores of the present invention are effective against a wide range of injurious agents, for example, but not limited to: oxidants, TNFα, neurotoxins, or radiation.
The zinc ionophores of the present invention are also effective in treating ischemia. Therefore, in a preferred form of treating ischemia the patient is administered a therapeutically effective amount of at least one zinc ionophore and a pharmaceutically acceptable carrier. A preferred zinc ionophore is zinc pyrithione. Another preferred zinc ionophore is zinc diethyldithiocarbamate.
Also defined within the present invention are compositions suitable for blocking apoptosis which comprise one or more zinc ionophores and a pharmaceutically acceptable carrier.
Various modifications may be made without departing from the invention. The disclosure is to be construed as exemplary, rather than limiting, and such changes within the principles of the invention as are obvious to one skilled in the art are intended to be included within the scope of the claims.
The present invention will now be demonstrated using specific examples that are not to be construed as limiting.
EXAMPLE 1 Screening for ionophores :
Cell Cultures:
Human umbilical vein endothelial cells (HUVEC) were purchased from Clonetics (San Diego, California) and passages 2-4 were used for these studies. Cells were cultured on flame- sterilzed glass coverslips in Endothelial Basal Medium (Clonetics) supplemented with lOng/ml human recombinant epidermal growth factor, 1.0 ug/ml hydrocortisone, 50ug/ml gentamicin, 50ng/ml amphotetericin B, 12ug/ml bovine brain extract and 2%v/v fetal bovine serum (all from Clonetics) , in a humidified chamber at 37°C and 5% C02. To maintain cell populations, proliferating HUVEC were passaged at 80-90% confluency. Cardiac myocytes were isolated from the ventricular septum of adult rabbit hearts, following collagenase digestion, in a manner similar to that described previously (Turan, B. et al., (1997) Am. J. Physiol. 272 :H2095-H2106) . The modification consisted of introducing low concentrations of CaCl2 during the perfusion with collagenase and the dispersion of the myocytes. Hearts were perfused for about 2 min by gravity under a hydrostatic pressure of 1 m, with a nominally Ca2+-free solution containing (in mM) : NaCl, 145; KC1, 5; MgS04, 1.2; Na2HP04 , 1.8; HEPES, 5; glucose, 10; pH adjusted to 7.4 with NaOH. Forty ml of this perfusate were then supplemented with collagenase (1 mg/ml) and perfusion was continued with recirculation. Within 2-3 min, this treatment resulted in a complete loss of ventricular pressure. The flow rate was then adjusted to 15 ml/min and 50 uM CaCl2 was added to the collagenase solution. Perfusion with this solution was continued for another 15 to 18 min, followed by a 2 min washout of the enzyme with fresh perfusate containing 100 μM CaCl2 and no collagenase. The hearts were then removed from the apparatus and the ventricular septum isolated and minced. Dissociation of the cells was obtained by gentle agitation of the minced tissue in 50 ml of the same perfusing solution. Following filtration through a 200 urn nylon mesh, the cells were allowed to settle and the supernatant was replaced with a solution containing 2 mM CaCl2. Cells were kept at 37°C in this pre-oxygenated solution and were studied within 8 hours after isolation, cellular viability was ensured by regularly replacing the incubation solution.
Primary cultures of mouse cerebellar granule neurons were obtained from dissociated cerebella of postnatal day 8 or 9 mice according to the following protocol (Cregan et al . , (1999) J. Neurosci . 19:7860- 7869, incorporated herein by reference). Brains were removed and placed into separate dishes containing solution A (124 mM NaCl, 5.37 mM KC1, 1 mM NaH2 P04 , 1.2 mM MgS04 , 14.5 mM D- (1) -glucose, 25 mM HEPES, 3 mg/ml BSA, pH 7.4) in which the cerebella were dissected, meninges removed, and tissue sliced into small pieces. The tissue was briefly centrifuged and transferred to solution A containing 0.25 mg/ml trypsin, then incubated at 37°C for 18 min. After the addition of 0.082 mg/ml trypsin inhibitor (Boehringer Mannheim, Indianapolis, I N) and 0.25 mg/ml DNase I (Boehringer Mannheim), the tissue was incubated at 25°C for 2 min. After a brief centrifugation, the resulting pellet was gently titrated in solution A yielding suspension that was further incubated for 10 min at 25°C in solution A containing 2.7 mM MgS04 and 0.03 mM CaC12. After a final centrifugation the pellet was resuspended in EMEM media (Sigma, St. Louis, MO) containing 10% dialyzed FBS (Sigma), 25 mM KC1, 2 mM glutamine (Life Technologies BRL, Gaithersburg, MD) , 25 mM glucose, and 0.1 mg/ml gentamycin (Sigma) and filtered through a cell strainer (size 70 μm; Falcon) . Cells were plated on glass coverslips coated with poly-D- lysine (Sigma) in Nunc four-well dishes at a density of 1.5xl05 cells per milliliter of medium. Cytosine- β- arabinoside (10 μM; Sigma) was added 24 hr after plating.
Test compounds:
Several test compounds with potential zinc- ionophore activity were screened for their ability to transport zinc into selected target cells. In order to ascertain that the transported ion was indeed Zn2+, and not some other divalent cation contaminant, the test compounds were first complexed with zinc. In addition to the zinc-complexed ionophores (holo- ionophores) , the zinc- free forms of these compounds (apo- ionophores) were also tested for the purpose of comparison. Whenever possible, purified holo- ionophores were purchased commercially (e.g. zinc-diethyldithiocarbamate, Sigma- Aldrich) . However, in most cases only the apo-ionophores were available commercially. The holo-ionophores were therefore prepared in our laboratory. Since zinc ionophores (e.g. pyrithione, diethyldithiocarbamate, 8- hydroxyquinoline) complex with zinc in a 2:1 molar ratio (ionophore: zinc) , stock solutions (generally 15.7 mM) of holo-ionophores were prepared by combining the apo- ionophore with ZnCl2 in a 2:1 molar ratio either in water or DMSO, depending on the solubility of the reactants, and incubating at room temperature for 15 min. The holo- ionophores were then stored at -20°C. Immediately prior to screening, the stock solutions of these test compounds were thawed and diluted in the superfusion buffer to give a final concentration of 1 μM of the holo- ionophore. When testing the apo-ionophores, an equivalent molar concentration of the ionophore in the superfusion buffer (2 uM) was used.
Ionophore screening:
Screening of the test compounds was performed with cultured HUVEC, isolated cardiac myocytes, and cultured cerebellar neurons following an approach described previously (Turan et al . , (1997) Am. J. Physiol. 272 :H2095 -H2106) . Immediately prior to screening, the cells were loaded with Fura-2, a zinc and calcium- sensitive indicator, by incubating the cells for 30 min in medium containing 4 uM Fura-2 -am (Molecular Probes) . Glass coverslips bearing HUVEC or cerebellar cells were placed directly in a superfusion chamber on the stage of an epifluorescence inverted microscope (Nikon Diaphot-DM) . With isolated myocytes, an aliquot of Fura-2 loaded cell suspension was placed in the superfusion chamber and the cells were allowed to adhere to the glass bottom of the chamber before superfusion was started. The microscope field of view was adjusted to include one or more individual cells. To establish baseline fluorescence, the cells were first superfused for a few minutes with a superfusing solution containing the following (in mM) : NaCl, 140; KC1, 5; MgCl2, 1; CaCl2, 2, HEPES, 5; glucose, 10; pH adjusted to 7.4 with NaOH. The flow rate was maintained at approximately 3 ml/min and the temperature at 37°C. The cells were then superfused with superfusion buffer containing a test compound and the fluorescence at 505 nm was recorded in response to excitation at 340 nm and 380 nm. The slope of the fluorescence intensity ratio in response to excitation at 340 and 380 nm was used to determine - ionophore activity. In each test, the membrane-permeant heavy metal chelator N,N,N' ,N' , - tetrakis (2 - pyridylmethyl) ethylenediamine (TPEN, 30 uM) was added to the superfusate at the end of the run. Since TPEN does not chelate Ca2+, loss of fluorescence in response to TPEN addition confirmed that the fluorescence was attributable to zinc. In cases where test holo-ionophores did not demonstrate zinc- ionophore activity, the validity of the negative observations was confirmed by adding zinc- pyrithione (1 uM) to the superfusing solution at the end of the test. An increase in fluorescence in response to the added zinc-pyrithione confirmed that the cell being tested was viable and responsive. Approximately 50 test compounds were screened for ionophore activity using this approach (See Table 1) . Of those, three groups of compounds were found to be particularly active zinc ionophores: pyrithione, dithiocarbamates, and hydroxyquinolines . Several compounds which do not belong to these groups also showed ionophore activity but at a lower level. The ionophore activity of pyrithione appeared to be comparable in all three cell types tested, as were the activities of diethyldithiocarbamate and 5, 7-diiodo- 8-hydroxyquinoline.
TABLE 1
EXAMPLE OF ZINC IONOPHORES
✓✓✓ - excellent
✓✓ - verv good
✓ - good
ZINC PYRITHIONE ✓✓✓
DITHIOCARBAMATES Pyrrolidinedithiocarbamate ✓✓ diethyldithiocarbamate ✓✓ Disulfiram ✓✓ dimethyldithiocarbamate ✓✓
HETEROCYCLIC AMINES 8-Hvdroxyσuinoline, Zinc salt ✓ 5 , 7 -Diiodo- 8 -hydroxyquinoline ✓✓
NSAID Indomethacin ✓
VITAMINS
Vitamin A (all - trans -retinol) ✓
Vitamin E (alpha- tocopherol) ✓
EXAMPLE 2 In vivo heart model - ischemic injury.
Experimental model: This model was designed to simulate myocardial infarcts in rats. The protocol is a modification of one described in detail previously (Fliss, H. et al., (1996) Circ. Res . 79:949-956, incorporated herein by reference) . Male Sprague-Dawley rats (250-350 g) were anesthetized, the chest was opened, a ligature was passed under the coronary artery and was then fashioned into a snare. The coronary artery was then occluded for a period of 45 minutes by tightening the snare, at which point the snare was released and the ischemic myocardium was reperfused. Following four hours of reperfusion the snare was re-tightened and the area- at-risk was delineated by intraveneous injection of the dye Evans blue. The rats were then killed immediately and the area-at-risk was collected for analysis. Previous studies have demonstrated that this model produces extensive myocardial apoptosis (Fliss, H. (1996) , supra) . To examine the protective ability of zinc-pyrithione in this model, different cumulative doses (from 0.9 mg/kg body weight to 1.2 ug/kg body weight) of this reagent in a 4% DMSO solution in sterile saline were injected intravenously through a tail vein in three equal boiuses : at the initiation of reperfusion, and at 1 and 2 h after the initiation of reperfusion. The total volume injected per rat was 1.5 ml . To examine the protective properties of zinc- diethyldithiocarbamate (ZnDDC) in this model, one dose of ZnDDC (0.21 ug/kg body weight) was tested in a manner identical to that used for zinc pyrithione. Results: The data collected to-date show a strong trend (P<0.1) towards anti -apoptotic effects with very low concentrations of zinc-pyrithione. TUNEL staining (Fliss, H. (1996), supra) , as described below, was used to identify the percent of apoptotic myocytes in the affected tissue. Parraffin sections were deparaffinized and were subsequently permeabilized with methanol/acetone (1:1) for 10 min at RT, and were washed twice with PBS. They were then incubated with 20 μg/ml proteinase K in 25 mmol/L Tris-HCl (1 ml/section), pH 6.6, for 15 min at RT, were washed twice (15 min each) with water, were stained with Hoechst 33258 (0.05 mg/ml) for 30 min at RT, protected from light, and were washed 3 times (1 min each) with PBS. The sections were then incubated in 75 ml of a buffer solution containing 200 mmol/L potassium cacodylate, 2 mmol/L CoCl2, 0.25 mg/ml bovine serum albumin, 25 mmol/L Tris-HCl, pH 6.6, 10 mmol/L biotin-16- dUTP (Boehringer Mannheim Canada, Laval, Quebec) , and 25 units of terminal transferase (Boehringer) , for 1 h at 37°C in a humidified chamber. The reaction was terminated by washing the sections 3 times (1 min each) with PBS at RT. The sections were then incubated with 1 ml of a staining solution containing 2.5 mg/ml fluorescein isothiocyanate-avidin (avidin-FITC) , 4X saline- sodium citrate buffer, 0.1% Triton X-100, and 5% powdered milk, for 30 min at RT, protected from light. The sections were washed 3 times with PBS, were coverslipped in "anti -fade" solution containing 1 mg/ml p-phenylenediamine, 90% glycerol, in PBS, and histofluorescence was monitored with a Zeiss Axiophot microscope. Positive control samples were prepared by incubating sections with 10 units/ml DNAse I for 20 min at 37°C prior to treatment with terminal transferase. The data demonstrate strong inhibition of apoptosis with 2.3 ug zinc-pyrithione per kg body weight when compared to DMSO carrier alone (P=0.067) (Figure 1). Similar strong protection against apoptosis was observed with 1.2ug/kg zinc-pyrithione (P=0.053). Since the zinc constitutes 20% by weight of zinc-pyrithione, the data suggest that about 0.20 to about 0.50 ug of zinc per kg body weight provides strong apoptosis protection. Strong protection (P<0.05) against apoptosis was also observed with the single dose of ZnDDC tested, i.e. 0.21 ug/kg body weight. Thus, the percent apoptotic nuclei in the ZnDDC treated hearts was 9.1+3.0, in comparison to the vehicle only group in which the percent apoptosis was 15.6±1.2.
Western blot analysis revealed that zinc- pyrithione (0.9 mg/kg body weight) significantly increased the mtracellular content of Heat Shock Protein 70 (HSP70) in the ischemic myocardium (Figure 2) . The content of the HSP70 in the ischemic myocardium was determined using standard immunoblotting techniques. The heart tissue was homogenized on ice for 45 s using a Polytron homogenizer at 10,000 rpm in 8 volumes of 10 mM HEPES (pH 7.9), containing 10 mM KC1, 1.5 mM MgCl2, 0.1% Nonidet P-40, 0.5 mM DTT, 0.5 mM PMSF, 0.5 mM spermidine, 0.15 mM spermine, 5 mg/ml aprotinin, 5 mg/ml leupeptin, and 5 mg/ml pepstatin. The homogenate was incubated on ice for 15 minutes and centrifuged at 35,000xg at 4°C for 15 min. Aliquots (15 mg protein) of the supernatant were subjected to electrophoresis on 12% polyacrylamide gels and were transferred to polyvinylidene difluoride (PVDF) membranes. The membrane was incubated with a polyclonal rabbit antibody against HSP70, followed by goat anti -rabbit IgG conjugated to horseradish peroxidase (HRP) . Protein band chemiluminescence was visualized on film according to manufacturer's instructions (NEN Life Science Products, Boston, MA) , and was quantified with a densitometer and Molecular Analyst Software (Bio-Rad Laboratories, Hercules, California) . Since HSP70 has been shown to protect against apoptosis (Wong, H.R., et al . (1996) Am . J. Respir. Cell Mol . Biol . 15:745-751), and to be induced by zinc (Klosterhalfen, B., et al . , (1997) Shock 7:254-262), the data suggest that zinc-pyrithione may exerts its protective effect by upregulating HSP70 synthesis. Zinc-pyrithione also caused a statistically significant decrease in the cytosolic concentration of PKC-α (data not shown) , using the same method as that described for HSP70, with the exception that instead of using a polyclonal rabbit antibody against HSP70, we used an antibody against PKC-α. As PKC-α is a well known mtracelular signalling agent, it was therefore concluded that Zn2+ is capable of modulating mtracellular signalling.
The limited amount of myocardial tissue available from these studies has not yet permitted the analysis of transcription factor activity in this tissue. However, analysis of brain tissue from the experimental rats by EMSA (electrophoretic mobility shift assays) showed strong effects on the nuclear content of the two transcription factors Spl and NF-kB. In the Electrophoretic Mobility Shift Assay (EMSA) , brain samples were homogenized on ice using six slow strokes of a Teflon pestle homogenizer at 1000 rpm in 8 volumes of buffer containing 0.25 M sucrose, 10 mM HEPES, pH 7.6, 25 mM KC1, 1 mM EDTA, 10% glycerol, 0.15 mM spermine, and 0.5 mM spermidine. The homogenate was filtered through a 45 mm nylon sieve and layered over a 10 ml cushion of 2 M sucrose containing 10 mM HEPES, pH 7.6 , 25 mM KC1, 1 mM
EDTA, and 10% glycerol. The homogenate was centrifuged at 100,000 xg at 4°C for 1 h, the supernatant was discarded, and the pelleted nuclei were gently resuspended in 40 ml of a lysis buffer containing 20 mM HEPES, pH 7.9, 420 mM NaCl, 1.5 mM MgCl2, 0.2 mM EDTA, 25% glycerol, 0.5 mM DTT, 0.5 mM PMSF, 0.5 mM spermidine, 0.15 mM spermine, and 5 mg/ml each of aprotinin, leupeptin and pepstatin. The suspension was incubated on ice for 45 min and centrifuged at 20,000xg at 4°C for 10 min. The supernatant containing nuclear protein was collected and diluted 1:1 with a buffer containing 20 mM HEPES, pH 7.9, 50 mM KC1, 0.2 mM EDTA, 20% glycerol, 0.5 mM DTT, 0.5 mM PMSF, 0.5 mM spermidine, 0.15 mM spermine, and 5 mg/ml each of aprotinin, leupeptin and pepstatin. Protein concentrations were determined using the Bio Rad protein assay. For EMSA assays, double- stranded consensus oligonucleotides for NF-kB, AP-1 and Spl (Promega, Madison, Wisconsin) were radiolabelled with g[32P]ATP (Amersham, Arlington Heights, Illinois) . Five mg of nuclear protein were first incubated for 10 min at room temperature with 5 mg poly-d[I-C] (Boehringer Manheim, Montreal, Quebec) in DNA binding buffer (20 mM HEPES, pH 7.9, 0.2 mM EDTA, 0.2 mM EGTA, 100 mM KC1, 5% glycerol, and 2 mM DTT) . Labeled probe (0.2 ng) was then added and the reaction mix incubated for a an additional 20 min in a final volume of 20 ml. The reaction mixture was subjected to electrophoresis on 5% polyacrylamide gel, and the dried gel was exposed to X-ray film. The intensity of the bands was quantitated with a densitometer and commercially available software (Molecular Analyst, Bio-Rad Laboratories, Hercules, California) . The subunit composition of NF-kB was determined with supershift assays. Antibodies (2 mg) to either p50 or p65 (Santa Cruz Biotechnologies, Inc. Santa Cruz, CA) were added to the incubation mixture and incubated for 20 min prior to the addition of poly-d[I- C] . No significant changes in the nuclear content of Spl were observed in control rats infused with zinc- pyrithione without coronary ligation, zinc-pyrithione strongly protected the brain tissue against the sharp decline in Spl caused by the ischemic episode (Figure 3) . Moreover, zinc-pyrithione significantly increased the level of NF-kB in the brain of rats subjected to myocardial ischemia, but had no effect on non- surgical control brains (data not shown) . In view of the well established role of these transcription factors in apoptosis, our data suggest that zinc-pyrithione may protect against apoptosis in the heart by altering cell signalling and gene expression.
EXAMPLE 3 In Vivo Brain model - kainic acid injury.
Glutamate is the principal excitatory neurotransmitter in the brain, and plays a critical role in the etiology of different major brain pathologies such as cerebral ischemia, neurodegeneration, epilepsy, etc (Coyle, J.T. (1993) Science 262 : 689 - 695) . Compounds which interact with the glutamate receptors are therefore important tools in the investigation of these diseases. Kainate is an excitotoxic glutamate analog that produces excessive neuronal excitation and seizures within hours following its intraperitoneal injection into adult rats. At 2-3 days after treatment, neurodegeneration can be observed in the limbic system in the form of apoptosis
(Gillardon, F., et al . , (1995) Neurosci . Let t . 192:85-88). Because the hippocampal subregions in the rat, particularly the CA3, are enriched in kainate receptors, they are particularly susceptible to kainate-induced neuronal death (Meldrum, B.S. (1994) Neurology
44:S14-S23). The neurotoxic effect of kainate in the rat hippocampal subregions involves a direct effect on presynaptic kainate receptors and an indirect effect on postsynaptic glutamate receptors due to the enhanced release of glutamate (Malva, J.O., et al . , (1998)
Neuroαhem . I fc . 32:1-6). It appears likely that kainate- induced apoptosis is associated with the production of reactive oxygen intermediates (Hirata, H. et al . , (1997) Brain Res .Mol . Brain Res . 48:145-148; Zhang, X., (1997) Eur. J. Neurosci . 9:760-769; Uz , T., et al . , (1996) Neuroscience 73:631-636; Rong, Y. et al . , (1996) J. Neurochem. 67:662-668), and the modulation of mtracellular zinc (Cuajungco, M.P. et al . , (1997) Neurobiol . Dis . 4:137-169).
Experimental model: The experimental approach we utilized in these studies was the well-established model of neurotoxic injury in rat brains induced by kainic acid (KA) . Male Wistar rats (250-350 g) were anethesized with chloral hydrate (325 mg/kg) . KA (10 mg/kg) was injected mtraperitoneally. Zinc pyrithione in 1.2% DMSO in sterile water was injected intracerebroventricularly according to the following coordinates: AP -6.8, L + - 1.5, DV +3.8 at the dose of 1 pmol/ventricle . Rats which did not receive zinc pyrithione were injected with 1.2% DMSO solution alone in water intracerebroventricularly. Zinc pyrithione was injected 15 min after KA administration. Sham operated rats received 1.2% DMSO in the lateral ventricles and isotonic saline mtraperitoneally. 'The volume of substances injected into ventricles was 1.5 uL. Seizure activity in the rats was followed during first 4-6 h after KA administration.
Gradation of KA- elicited limbic seizures was carried out according to accepted protocols (Rong, Y. et al . , (1996) J. Neurochem . 67:662-668). Sham operated, KA- treated and KA/zinc treated rats were decapitated after 1-3 days. The brains were removed, fixed in AFA, paraffin embedded, sectioned (10 μm) and stained by the Nissl method, using routine methodology.
Results: Brains of sham-operated rats and control animals injected icv with zinc-pyrithione did not show any obvious damage in any brain region. However, administration of KA caused neuronal degeneration and cell loss in a number of brain regions, with injury reaching maximal levels within the first day after KA treatment. Preliminary data suggest that the brain damage is attributable to apoptosis, as detected by the TUNEL stain (not shown) . Twelve rats were subjected to KA treatment alone and 12 were treated with KA followed by zinc-pyrithione. Figure 6 shows the number of rats (out of 12) that displayed detectable signs of injury in each group. The extent of injury in tissue sections was determined by thorough histological examination, comparing gross anatomical features, the number of visible, intact nuclei in each region, and the presence of other obvious signs of tissue injury. Reproducible and pronounced damage was seen in the hippocampal subregions CAI and CA3 , the pyriform cortex (PC), amygdalar region (AM) , and thalamus (TL) in the KA- treated group (Figure 6) . However, icv administration of zinc-pyrithione provided statistically significant protection in all regions with the exception of CA3 , where only a protective trend was observed (*, P<0.1
(trend); **, P<0.05; ***, P<0.01, Fisher's exact test).
Zinc-pyrithione also changed the pattern of KA- induced seizures in the rats. The seizure study was performed with 57 rats in the KA- alone group, and 58 in the KA plus zinc-pyrithione group. The data presented in Figure 5 show the number of rats in which a given seizure severity was the final stage of severity observed. Notably, zinc-pyrithione caused a statistically significant 3.2 -fold decrease in the incidence of the most severe and irreversible stage of seizures
("jumping", stage 6). In other words, of the 57 rats treated with KA alone, fully 18 reached the lethal stage 6, as compared with only 5 out of 58 in the KA plus zinc- pyrithione group. The data further show that the final stage of severity tended to be much lower in the zinc- pyrithione group, as illustrated by the 3.2-fold increase in the number of zinc-pyrithione treated rats at level 2 ("wet dog shake" stage). (*, P<0.01, Fisher's exact test) . In summary, zinc-pyrithione significantly decreased KA- induced cell death in a number of brain regions, and significantly lowered the severity of KA- induced seizures in rats.
EXAMPLE 4 In vi tro neuronal cell model - oxidative stress
Oxidative stress is believed to play an important role in the apoptotic neuronal cell death associated with many different neurodegenerative conditions (e.g., Alzheimer's disease, Parkinson's disease, cerebral ischemia, etc.) (Jenner, P. (1994) Lancet 344:796-798). The non-differentiated rat pheochromocytoma PC12 cells are a cell line which differentiates to a neuronal cell type in the presence of Nerve Growth Factor (NGF) , but undergoes apoptotic cell death when deprived of NGF. These cells also undergo apoptotic cell death when exposed to oxidants such as hydrogen peroxide (Satoh, T., et al . , (1997) J. Neurosci . Res . 50:413-420; Maroto, R. et al . , (1997)
J. Neurochem . 69:514-523; Kubo, T., et al . , (1996) Brain Res . 733:175-183). PC12 cells are therefore a useful cell model with which to analyze the molecular mechanisms of apoptosis induced by oxidative stress and other stimuli in neuronal cells (Kubo, T.,et al . , (1996) Brain Res . 733:175-183, incorporated herein by reference).
Experimental model: PC12 cells were seeded on coverslips covered with poly-L- lysine in 24 -well plates, and were grown in RPMI-1640 containing 10% FCS, 5% horse serum, 2 mM glutamine, and 40 mg/kg gentamicin in 5% C02 at 37°C. RPMI-1640 medium containing 1% serum (embryonal calf and horse serum, 2:1) and 50 ng/ml NGF was used to induce differentiation. Differentiated PC12 monolayers were washed and were induced to undergo apoptosis in two ways: a) incubation for 4 h with normal growth medium without serum or NGF, and b) incubation for 4 h with normal growth medium (plus serum and NGF) containing 20 μM hydrogen peroxide (H202) . The protective effect of zinc- pyrithione against both types of apoptosis was tested by preincubating the cells with this compound for only 5 minutes immediately prior to initiating apoptosis - inducing treatments a or b. Control cells were pretreated with the carrier DMSO alone. Pretreatment with zinc-pyrithione, rather than the more relevant post- treatment approach, was used in this model because of practical experimental considerations. However, the close temporal proximity of the pretreatment to the initiation of the injurious treatment is more representative of a concomitant exposure of the cells to both zinc-pyrithione and the injurious agent, rather than an authentic pretreatment. At the end of the 4 h incubation, the cells were fixed with methanol : acetone (1:1) at -20°C and were stained with Hoechst 33258 to> visualize the nuclei.
Results: The data show that zinc-pyrithione (2-500 nM) provided statistically significant protection against H202- induced apoptosis, with 10 nM being the optimal concentration (Figure 6) . Compared to control cells (+NGF) , treatment with H202 resulted in the loss of approximately 50% of the cells through apoptosis (+NGF, +H202) . However, treatment with as little as 2 nM zinc- pyrithione for 5 min significantly attenuated cell death. (*, P<0.005 vs. +NGF; +, P<0.05 vs. NGF+H202 ; ++, P<0.02 vs. NGF+H202 , Mann-Whitney test, n=4). In another series of tests (Figure 7) , the ability of zinc-pyrithione to protect against H202 was confirmed, and also demonstrated that this compound can block the apoptosis caused by serum and NGF deprivation. Here too 10 nM appeared to be the optimally protective concentration of zinc-pyrithione. Zinc-pyrithione at concentrations as high as 1000 nM did not show any effects on cell growth in control cultures. (*, P<0.05 vs. +NGF; +, P<0.05 vs. respective "without ZP" group, Mann-Whitney test, n=3) . The addition of Zn2+ alone, at concentrations equivalent to those of zinc-pyrithione, to H202-treated cells did not block apoptosis (not shown), suggesting that the ionophore pyrithione is necessary for the transport of Zn2+ into the cytosol, and that it is this mtracellular Zn2+ which accounts for the protective effects of zinc-pyrithione.
EXAMPLE 5 In vi tro endothelial model - response to ionizing radiation and TNFα
Ionizing radiation has been shown to induce apoptosis in a variety of cell and tissue types (Van
Antwerp, D.J., et al . (1996) Science 274:787-789; Findik, D., et al., (1995) J. Cell . Biochem . 57:12-21) including endothelial cells (Langley, R.E., et al . (1997) Br. J. Cancer 75:666-672). Although the mechanisms by which ionizing radiation induces apoptosis have yet to be resolved, it has been shown to directly induce DNA damage, to generate the formation of reactive oxygen species and to alter membrane structure (Datta, R. , et al. (1997) J. Biol . Chem . 272:1965-1969), all of which can contribute to apoptotic cell death. One well established effect of ionizing radiation is its alteration of gene transcription by means of the modulation of transcription factors. For example, radiation has been shown to induce the activation of the transcription factor NF-kB in several cell types (Valerie, K. , et al . , (1995) Biochemistry 34:15768-15776), including endothelial cells (Hallahan, D. et al . , (1995) Biochem . Biophys . Res . Commun . 217:784-795). The cytokine TNFα is also capable of causing apoptosis in endothelial cells either alone
(Slowik, M.R., et al . , (1997) Lab. Invest. 77:257-267; Spyridopoulos, I., et al . , (1998) Circula tion 98:2883-2890) or synergistically with other agents (Eissner, G., et al . , (1995) Blood 86 :4184-4193) . Here too, the apoptotic events are heavily regulated by alterations in transcription factor activity (Hu, X.L., (1998) Blood 92:2759-2765). The studies performed with this model were designed to examine the ability of zinc- pyrithione to block radiation- induced apoptosis, and to elucidate the effect of zinc-pyrithione at the transcriptional level in response to either ionizing radiation or TNFα.
Experimental model :
Cell Culture: Human umbilical vein endothelial cells (HUVEC) were purchased from Clonetics (San Diego, California) and used from passages 2-4. Cells were cultured on gelatin- coated culture dishes in Endothelial Basal Medium (Clonetics) supplemented with lOng/ml human recombinant epidermal growth factor, 1.0 ug/ml hydrocortisone, 50ug/ml gentamicin, 50ng/ml amphotetericin B, 12ug/ml bovine brain extract and 2%v/v fetal bovine serum, in a humidified chamber at 37°C and 5% C02. To maintain cell populations, proliferating HUVEC were passaged at 80-90% confluency.
Experimental Treatments: HUVEC were grown to confluency, and then given an additional 24 hours to achieve quiescence prior to experimental treatment. The following treatments were performed:
Radiation: The cells were washed twice with 37°C D-PBS and then irradiated in fresh media. Irradiated cells received a dose of 1000 Rads of gamma- irradiation from a 137Cesium source (Atomic Energy of Canada) . The cells were then incubated for 2 hours (cytosolic and nuclear protein extraction) or 8 hours (Hoechst staining and DNA electrophoresis) . TNFα: The cells were washed twice with 37°C D-PBS and then incubated in media containing the TNFα (20ng/ml, from a stock of lOug/ml prepared in phosphate buffered saline (PBS) - l%bovine serum albumin. Control cells received fresh media alone. The cells were then incubated for 2 hours (cytosolic and nuclear protein extraction) or 8 hours (Hoechst staining and DNA electrophoresis) .
Hoechst Staining: Cells were grown on round, gelatin coated 12 mm glass coverslips, and following treatment, were fixed with 0.5 ml of 1% glutaraldehyde in PBS for 10 minutes at room temperature (RT) . The cells were then washed twice with PBS for 5 minutes, and permeabilized with 0.5 ml of 1:1 methanol/acetone for 10 minutes at RT, followed by two five minute PBS washes. The cells were then incubated with Hoechst 33258 (bis-benzimide, 0.05mg/ml in H20) , a fluorescent DNA binding dye, for 30 minutes at room temperature, in the dark. The nuclear morphology of the cells was then visualized under a Zeiss Axiophot fluorescence microscope.
Preparation of Cytosolic and Nuclear Extracts: Cells were grown on 100mm2 culture dishes, and following treatment, were scraped into ice cold PBS and collected by centrifugation at 200Xg for 5 minutes. The cells were then resuspended and washed once in 1 ml of ice cold PBS and centrifuged at 200Xg for 5 minutes at 4°C. The cells were resuspended in 1 ml of Buffer A (lO M HEPES, lOmM KC1, 1.5mM MgC12, pH=7.9, 1.5mM DTT and 0.5mM phenyl methyl sulphonyl fluoride (PMSF) ) and centrifuged at 200Xg for 5 minutes at 4°C. The cells were then resuspended and lysed in 300ul of Buffer A containing 0.1% Nonidet P-40 for 25 minutes on ice. The homogenate was then spun at 20,000Xg for 10 minutes at 4°C. The supernatant containing cytosolic proteins was combined with an equal volume of Buffer C (20mM HEPES, 50mM KC1 , 1.0 mM EDTA, 0. ImM EGTA, 20% glycerol, pH=7.9, 0.5mM DTT and 0.5mM PMSF) and was stored at -80°C. The pelleted nuclei were washed by resuspension in 1ml of Buffer A and spun at 20,000Xg for 1 minute. The supernatant containing residual cytosolic proteins was discarded and the pelleted nuclei were resuspended in 35ul of Buffer B (20mM HEPES, 420mM NaCl, 1.5mM MgCl2, 0.2mM EDTA, 25% glycerol, pH 7.9, 0.5mM DTT, 0.5mM PMSF, and the protease inhibitors spermidine, spermine, aprotinin, leupeptin and pepstatin) for 45 minutes on ice in order to extract the nuclear proteins. The nuclear extract was then obtained following centrifugation at 20,000Xg for 15 minutes at 4°C, and was combined with an equal volume of Buffer C and stored at -80°C.
Determination of Protein Concentration: The protein concentration in the nuclear and cytosolic extracts was determined using the Bradford Assay (Biorad) using bovine serum albumin as the standard.
Electrophoretic Mobility Shift Assay (EMSA) : Equal amounts of nuclear protein (5ug) were incubated with poly dl-dC (5ug from a stock of 2.5ug/ul in TE buffer) for 10 minutes at RT. This reaction mixture was then incubated with 0.2ng of 5' end-32phosphorus - labelled double stranded oligonucleotide probe for 20 minutes at RT to allow the binding of nuclear proteins with the labeled probe. Loading buffer (5ul of a mixture containing 20mM HEPES, lOOmM KC1, 60% glycerol, 0.5mM EDTA, 0.5mM EGTA and 0.125% bromophenol blue) was added to the reaction mixture prior to the electrophoresis on a 5% native polyacrylamide gel. The gels were run in Tris-Glycine solution for 1.5 hours at 200V and were then dried between filter paper and cellophane for 1.5 hours at 80°C under vacuum. The dried gels were exposed to X-ray film (Cronex) for up to 2 days at -80°C. For competition assays, the reaction mixture was incubated with a 125- fold excess of unlabeled probe for 20 minutes at RT prior to the addition of the labeled probe. For supershift assays, the reaction mixture was incubated with 2mg of rabbit polyclonal anti-NFkB p50 or p65 antibody (Santa Cruz Biotechnology) for 20 minutes at RT immediately subsequent to the addition of the labeled probe. The bound antibody retards the mobility of the protein-DNA complex, resulting in a shifted band. The consensus oligonucleotides for the transcription factors NFkB (5' - ACT TGA GGG GAC TTT CCC AGG C - 3 ' ) , AP - 1 ( 5 ' - CGC TTG ATG AGT CAG CCG GAA-3') and Spl (5' -ATT CGA TCG GGG CGG GGC GAG C-3') (Promega) and were labeled as suggested by Promega with minor modifications. Briefly, oligonucleotides (20ng) , T4 Polynucleotide kinase and [g32P]ATP (60uCi) were mixed in kinase buffer (50mM Tris- HCl, pH 7.6, lOmM MgCl2, 5% glycerol and 5mM DTT) and incubated at 37°C for 1 hour. Labeled oligonucleotides were removed by centrifugation through a G-25 Sephadex Column at 8500rpm for 20 minutes. The labeled oligonucleotides were then diluted such that 2ul of the probe mixture contained approximately 50000 - lOOOOOcpm. Western Blotting: Equal amounts of cytosolic protein (3ug) were diluted 1:1 in sample buffer (0.125M Tris-HCl pH 6.8, 2.6%SDS, 25% glycerol, 0.1ml beta-mercaptoethanol and bromo phenol blue) . The mixture was placed in boiling water for 5 minutes to denature the proteins and was then subjected to SDS -PAGE for 2.5 hours at 100V in running buffer. The gels consisted of a stacking gel (4.5% acrylamide, 0.125M Tris-HCl pH=6.8, 0.1% SDS, 0.6% ammonium persulfate and 0.2% TEMED in H20) and a 10% running gel (10% acrylamide, 0.3% bis acrylamide, 8% glycerol, 0.375 Tris-HCl pH=8.8, 0.1% SDS, 0.04% ammonium persulfate and 0.05% TEMED in H20) . After electrophoresis (100V, 50 minutes) , the gels were equilibrated for 15 minutes in ice cold transfer buffer (25mM Tris HC1, 20% methanol, and 192mM glycine) , then transferred onto a polyvinyllidene difluoride membrane for 1 hour at 100V. The blots were then blocked overnight in 5% skimmed milk in Tris-Buffered Saline containing 0.1% Tween-20 (TBS-T) at 4°C with constant shaking. The blots were then washed with TBS-T and incubated for 1.5 hours in primary antibody (anti-IkBa, Santa Cruz Biotechnology) diluted 1:1000 in 2% skimmed milk in TBS-T and sodium azide at RT with constant shaking. The blots were then washed with TBS-T and incubated for 30 minutes in horseradish peroxidase labeled goat anti -rabbit IgG diluted 1:10000 in 2% skimmed milk in TBS-T at RT with constant shaking. Following treatment with the secondary antibody, the blots were extensively washed with TBS-T and incubated for 1 minute with chemiluminescent substrate. The blots were then exposed to X-ray film for 1-5 minutes. Results: The data show protection by zinc-pyrithione against endothelial apoptosis, and also show that this protective effect is associated with transcriptional modulation. Treatment of irradiated HUVEC (IR) with zinc-pyrithione significantly blocked apoptosis (Figure 8) . The data also show that zinc is required for this effect since the sodium salt of pyrithione was not effective in preventing apoptosis. DMSO alone was also ineffective. No apoptosis was caused in control cultures by zinc-pyrithione, sodium-pyrithione, or DMSO alone (Figure 8). (*, P<0.05 vs. Control; °, P<0.05 vs. irradiated cells, n=4).
EMSA tests showed that irradiation- induced apoptosis is associated with a significant increase in nuclear NF-kB content, and that zinc-pyrithione, but not sodium-pyrithione or DMSO alone, blocked this increase (Figure 9) . Zinc-pyrithione also lowered the nuclear content of AP-1, but did not appear to affect Spl in this model. (*, P<0.05 vs. Control; °, P<0.05 vs. irradiated group, n=4) . Zinc-pyrithione had a very similar effect in TNFα-treated HUVEC (Figure 10), and was particularly potent at blocking the TNF -induced increase in NF-kB content. (*, °, same as above, n=3) . Since NF-kB is associated with the cytosolic inhibitor IkB which governs its activity in the cell, the effect of zinc-pyrithione on the cytosolic level of this protein was examined. The data show that zinc-pyrithione lowered the cytosolic content of IkB in cells treated with either radiation or TNFα (Figure 11) .
EXAMPLE 6 EFFECTS OF ZINC PYRITHIONE ON MODELS OF ISCHEMIC STROKE IN RODENTS
Stroke is an extremely variable clinical condition which reflects the variability of the underlying disease process. The vascular occlusion can occur at many different sites in the brain and the cause of the occlusion, the severity of the problem, and the degree of reversibility can all contribute to the variability of outcome. In contrast, in experimental animal models most of these variables can be controlled or eliminated, enabling a meaningful interpretation of the results. Generally, stroke models are grouped into those producing either global or focal ischemia
(Ginsberg, M.D. & Busto, R. (1989) Stroke, 20:1627-1642, incoporated herein by reference; Ginsberg, M.D. & Busto, R. (1998) Small -Animal Models of global and focal cerebral ischemia. In Cerebrovascular Disease: Pathophysiology, Diagnosis, and Management (ed. Maiden, M.A.), pp. 14-35, Blackwell Science, incoporated herein by reference) . It is generally understood that global models are more relevant to cardiac arrest, while focal models are of greater relevance to acute ischemic stroke. To study the possible neuroprotective effects of zinc pyrithione (ZP) two models of stroke were used: a global ischemia -reperfusion model of 4 vessel occlusion in rats (4VO) and a focal ischemia model of middle cerebral artery occlusion in mice (MCAO) . The 4VO approach sealed off the two carotid and two vertebral arteries which carry all the blood to the brain. This approach permitted severe forebrain ischemia to be produced in awake and freely moving rats, and produced reproducible neuropathology. The 4VO is a two-stage operative procedure (Pulsinelli, et al . , (1979) Stroke, 10: 267-272, incorporated herein by reference) . In the first stage, the vertebral arteries were exposed and permanently sealed by electrocauterization (Pulsinelli, et al., (1988) Stroke, 19:913-914, incorporated herein by reference) . This occlusion of the vertebral arteries does not in itself cause serious injury in the rat. It is the second stage which initiated the injurious ischemic episode. It was performed 24 h later and involved the brief occlusion of the carotid arteries, shutting off all blood flow to the brain. Although there was a marked mortality during both stages even in laboratories which are highly experienced in this procedure (Ginsberg, et al . , (1989) supra) the 4VO is a favorite stroke model because it results in highly reproducible damage in the CAI region of the hippocampus, as well as in some other brain regions. MCAO in mice is one of the most clinically relevant stroke models. It shuts off blood flow to only a portion of the brain, producing a focal injury which closely resembles the clinical situation with stroke patients. This procedure was performed with an intraluminal thread. A nylon suture was introduced into the external carotid artery and was gently advanced into the internal carotid artery. The diameter of the suture was such that it lodged in the anterior cerebral artery, occluding the medial cerebral artery at its origin. Brain damage in this model was observed as early as several hours after the ischemic episode, with optimal injury occurring at 24 h. The injury occupied a large part of the hemisphere including the cerebral cortex and subcortical structures, and its severity depended on the duration of ischemia and the strain of mice used.
Animals
The 4VO procedure was performed on male Wistar rats weighing 240-300 g. The rats were housed in groups of 5 in plastic cages with free access to food and water. The MCAO protocol was performed on male C57BL/6 mice weighing 20-28 g which were housed in groups of 10 in plastic cages with free access to food and water. All experiments were performed in accordance with the National Institutes of Health Guidelines for the Care and Use of Laboratory Animals.
Surgeri es 4VO in rats
Rats under chloral hydrate anesthesia (325 mg/kg) were positioned in a stereotaxic frame. The vertebral arteries were exposed and permanently occluded by electrocautery at the first cervical vertebra. Snares (surgical silk strings) were then placed loosely around each common carotid artery without interrupting the carotid blood flow. The animals were then allowed to recover for 24 hours with free access to water. On the following day, the rats were lightly anesthetized with ether, were secured to surgical boards, ventral side up, and their common carotid arteries were exposed. Forebrain ischemia was initiated by tightening the snares around the carotid arteries for 10 min. The body temperature of the rats was carefully maintained at ~ 37-37.5°C, both before and during the ischemic insult, using a feedback- controlled heating pad and a rectal thermistor (Homeothermic Blanket System, Harvard Apparatus LTD, England) . The initial (1st min of ischemia) and final (10th minute of ischemia) temperature did not differ among all groups of rats studied. After the ischemic episode, the temperature was maintained in similar fashion at 37°C for at least 4 h. Only rats that showed signs of severe neurological injury, such as a loss of the righting reflex, pupil dilation, etc., were included in this study. In the sham-operated controls, the vertebral and carotid arteries were exposed, but were not occluded. Evaluations of neurological deficit were performed at 24 and 96 h after ischemia, and were based on a scoring system which recorded activity level, motility, pain reflex, grabbing reflex, and the ability to see and hear (Miljkovic, L.M., et al . , (1997) Ann. Emerg. Med. 29, 758-765, incorporated herein by reference).
MCAO in mice Mice (C57BL/6) were anesthetized with an intraperitoneal injection of chloral hydrate (350 mg/kg) and xylasine (4 mg/kg) . Focal cerebral ischemia was produced by occlusion of the MCA using the intraluminal filament technique (Longa, Z.E., et al . , (1989) Stroke 20 : 84 -91, incorporated herein by reference). A 8.0 nylon microfilament coated with a silicon resin (Xantopren) - hardener mixture (Hara, H., et al . , (1996) J. Cereb. Blood Flow Metab. 16:605-611, incorporated herein by reference) was inserted into the left common carotid artery, and was advanced 10-11 mm distal to the carotid bifurcation so as to occlude the MCA and posterior communicating artery. The filament was left in this position for 1 h. For reperfusion, the animals were re- anesthetized briefly and the filament was withdrawn to restore the blood flow. Core temperature was maintained at ~37°C with a homeothermic blanket for a period of 2 h following reperfusion. The neurological deficit caused by the ischemic insult was scored after 2 h of reperfusion according to the scheme of Bederson, J.B., et al . (1986) Stroke 17:472-476, incorporated herein by reference: 0, no observable neurological deficit (normal); 1, failure to extend the right forepaw (mild); 2, circling to the contralateral side (moderate); 3, falling to the right (severe); 4, inability to walk spontaneously (most severe) .
Zinc ionophore treatments
The zinc pyrithione (ZP) data presented were derived with a treatment protocol in which ZP was injected in three boluses, at 10 min, 1 h, and 2 h after the termination of the ischemic episode, through a tail vein catheter. With the 4VO rat model, four doses of ZP: 3x1.2 μg/kg, 3x6 μg/kg, 3x30 μg/kg, and 3x200 μg/kg were tested. With the mouse MCAO model only the three lowest concentrations of ZP were tested. Some data were also obtained with a second ZP treatment protocol in which three boluses of 6 μg/kg (3x6 μg/kg) were injected at 3, 4, and 6 h after the termination of the ischemic episode. In addition, a treatment protocol with zinc- diethyldithiocarbamate (ZnDDC) was performed with both the 4VO and MCAO models using the dose of 3x7.6 μg/kg, a regimen which delivers zinc at a dose equivalent to that delivered with 3x6 μg/kg of zinc pyrithione. The zinc ionophore solutions were prepared by diluting a stock solution of ZP and ZnDDC in DMSO with saline. The final concentration of DMSO in the injectate was 2.5%. Control animals receiving vehicle- alone were injected with 2.5% DMSO in saline.
Histology 4VO
Four days post surgery, control and experimental animals were deeply anaesthetized with sodium thiopental (60 mg/kg, mtraperitoneally) and were perfused transcardially with 250 ml of AFA fixative (96% alcohol, 39 % formalin, glacial acetic acid , 7:2:1). After the AFA perfusion the heads were collected intact and were kept at 4°C for 4-5 h. The brains were then removed and immersed in the same fixative for 1 h, and were then stored in 70% alcohol. Each forebrain was cut into three frontal blocks and imbedded in paraffin. Ten μm thick sections were cut from a region 3.0-4.0 mm posterior to the bregma. The sections were stained with cresyl violet (Nissl) . Computer images of the stained sections were prepared, and the total number of viable pyramidal neurons was counted in a 500 um-long section of the CAI region in the hippocampus . The person doing the cell counts was blinded as to the identity of the experimental groups. In Situ End Labeling (ISEL) , a protocol for identifying apoptotic cells by staining fragmented DNA, was performed according to a protocol developed in the laboratory of Dr. Fliss (Schmidt - Kastner, et al . , (1997) Stroke 28: 163-170, incorporated herein by reference) using deparaffinized 10 μm thick brain sections from rats sacrificed at 24 h and 96 h after ischemia. Fluorescence was monitored with a Zeiss Axioplan microscope.
MCAO Mice were killed 24 h after reperfusion with an overdose of sodium thiopental (60 mg/kg, mtraperitoneally) , and the brain was rapidly removed and sectioned coronally into five 1.7 mm slices. The slices were then placed in 2% (wt/vol) 2,3,5- triphenyltetrazolium chloride solution (TTC) in PBS (pH 7.4) for 20 min at 37°C. This procedure, which tests mitochondrial activity, stains viable tissue a bright red, while the infarcted regions remain white. Following TTC staining the sections were fixed in 10% formalin overnight. The area of infarct in each section was determined using an image-analysis system. The infarct volume was subsequently calculated by summing the infarct areas in the sequential 1.7 mm- thick sections with correction for edema. The person measuring infarct volumes was blinded as to the identity of the experimental groups. For ISEL staining, the brains were removed, were frozen rapidly, and were sectioned with a cryostat into 20 urn sections from the anterior side to the posterior side at 500 urn intervals. ISEL was performed as described above. Adjacent sections were stained with cresyl violet (Nissl) . The ApopTag® Peroxidase In situ Apoptosis detection Kit (Intergen) was also used to detect apoptosis in mouse brain sections .
Statistics
The data were expressed as means ±S.E.M. One way- ANOVA with post hoc Duncan's test or T-test for independent samples were used for statistical analysis of data. Zinc ionophore toxic ity
No noticeable changes in behavior or appearance were detected in animals injected with lower doses of ZP, or the single dose of ZnDDC, when compared to those receiving vehicle alone. The weight loss in rats after 4VO (7.5-16.9 % at 24 h after ischemia and 4.1-11.2% at 96 h after ischemia) did not differ between the vehicle- treated and ZP- treated groups.
4VO model (10 min - 1 h - 2 h - injection schedule) Neuronal cell loss
Approximately 15% of the neurons in the CAI region survived the ischemic insult in the vehicle- treated group when compared to the sham-operated animals. However, treatment with ZP showed pronounced protection and increased the number of viable cells (Figure 12) . The ZP- mediated increase in cell survival varied (1.6-3.5 fold) and did not show a clear dose-dependence (Figure 13) . Although all doses of ZP tested showed evidence of protection, the 3x6 μg/kg group reached statistical significance with approximately 52% of the pyramidal cells surviving the ischemic episode. One-way ANOVA (sham, vehicle, and ZP- treated groups) showed a significant dependence of the viable cell number in the CAI on the experimental treatment conditions (F=12.9, P<0.000001). The post hoc Duncan's test showed the difference of all ZP and vehicle- treated groups from shams at P<0.01, with the ZP 3x6 μg/kg group significantly different from vehicle at P<0.01, and the 3x200 μg/kg ZP group showing a trend at P<0.09. Thus, 3x6 μg/kg appeared to be the optimal protective dose of ZP in this model. However, the trend for significant protection shown by the 3x200 μg/kg ZP group, and the collective evidence of protection at the other doses suggested that ZP is protective over a broad range of doses .
In addition, with ZnDDC at 3x7.6 μg/kg, the number of viable neurons in the CAI region was 99.5± 19.9, showing that the protection with this dose of ZnDDC was similar to that achieved with 3x6 μg/kg of ZP, a dose which delivered the same amount of zinc (Figure 13A) . ISEL staining As described above, ZP at 3x6 μg/kg showed significant protection against neuronal cell death. However, the data do not indicate if this cell death was apoptotic or necrotic in nature. To determine if the ZP- dependent increase in cell survival was attributable to a lower incidence of apoptosis we performed ISEL on brain sections .
At 24 h after ischemia no apoptotic cells were observed in the sham, vehicle- treated, or ZP (3x6 μg/kg) - treated groups (n=5 for each group) , suggesting that apoptosis required a longer post- ischemic period to manifest itself in this model. Apoptosis was more commonly observed at 96 h after ischemia. Numerous ISEL- positive nuclei were observed in the hippocampi of vehicle- treated rats (131.4 ±14.6 in CAI region, n=5). However, in rats treated with 3x6 μg/kg ZP, the number of apoptotic nuclei was approximately 4 times lower (33.6±7.8, n=9, P<0.0001, t-test) (Figure 14). No apoptosis was detected in any brain region of the sham- operated rats. The data therefore indicated that ZP has a potent anti-apoptotic effect in this model, and that it is this effect which accounted for the observed neuronal protection.
Neurological defici t
The neurological deficit data in 4VO rats are presented in Table 2. A method of scoring in which increasing neurological scores are indicative of decreasing neurological function compared to a perfect score of 0 for the shams was employed. For example, a large number of the 4VO rats did not show any neurological deficit (score 0) , despite the fact that they sustained an almost complete loss of cells in the CAl region. Therefore, the administration of ZP did not influence the neurological deficit in the 4VO rats despite clear evidence of histological protection.
4VO model (3 h, 4 h, 6 h injection schedule)
Administration of ZP in boluses of 6 μg/ g at 3, 4, and 6 h after the ischemic episode (3x6 μg/kg) significantly increased neuronal viability in the CAl region, with the number of viable neurons increasing 2.5 fold vs. the vehicle- treated group. The cell count was 73.2+15.0 in the ZP- treated group vs. 29.2±7.8 in the vehicle- treated group (P<0.03, t-test). The viable cell counts in the 3x6 μg/kg groups of both injection regimens (10 min, 1 h, 2 h or 3 , 4 , and 6 h after ischemic episode) did not differ significantly (103.5±15.2 vs. 73.2±15.0). The neurological deficit data for this ZP administration schedule are presented in Table 2. The neurological score in "3, 4, 6 h schedule" rats did not differ from that in the other regimen (10 min, 1 h, 2 h after ischemic episode) . These data therefore indicated that delaying the first administration of ZP by 3 hours did not significantly change its neuroprotective effect at the dose of 3x6 μg/kg, indicating a possible wide therapeutic window for ZP in the 4VO stroke model.
MCAO MCAO for 1 h produced significant infarcts in the left hemisphere of mouse brain (Figure 15) . Three doses of ZP were used (3x1.2 μg/kg, 3x6 μg/kg, and 3x30 μg/kg) and infarct areas, infarct volumes and neurological scores were measured. ZP at all three doses significantly decreased the infarct area at a distance of 3.4-5.8 mm from the frontal pole (Figure 16) . One-way ANOVA (vehicle and ZP- treated groups) showed a dependence of infarct size in sections 3, 4, and 5 (3.4, 4.1, and 5.8 mm from the frontal pole) on the experimental treatments with F=7.8 (P=0.0003), 13.0 (P=0.000003) , and 11.7
(P=0.00001), respectively. The post hoc Duncan's test showed a significant difference between infarct size and vehicle at P<0.01 in sections 3-5 for each ZP dose. The infarct size did not differ significantly between the ZP groups. In addition, ZnDDC at 3x7.6 μg/kg also significantly decreased the infarct area in this model (Figure 16A) .
Table 3 presents data on the absolute and relative (% of the contralateral hemisphere) infarct volumes in the mouse MCAO model in response to zinc pyrithione treatment. The calculated infarct volumes in the ischemic control mice were similar to those reported previously for this strain of mouse (Hara, H. , et al . (1996) J. Cereb. Blood Flow Metab. 16:605-611; MacManus, J.P., et al., (1999) NeuroReport 10:2711-2714; Nagayama, M., et al., (1999) J Cereb. Blood Flow Metab. 11:1213- 1219; Nogawa, S., et al . , (1998) Proc . Natl . Acad. Sci. U S A. 95: 10966-10971; Takagi, Y. , et al . , (1999) Proc. Natl. Acad. Sci. U S A. 96:4131-4136). Vehicle alone (2.5% DMSO) produced apparently contradictory effects. It tended to increase the infarct volume but concomitantly decreased the neurological score. However, these effects were not statistically significant.
The administration of ZP at all three doses statistically significantly decreased both the absolute (data not shown) and relative infarct volumes by 29.0- 38.2% and 30.8-40.0%, depending on the dose, respectively) compared with the vehicle- treated group (Figure 17). Comparison of control, vehicle-, and ZP- treated groups using one-way ANOVA showed a significant dependence of both absolute and relative infarct volumes on the experimental treatments (F=10.1, P<0.00005; F=8.2, P<0.00005, respectively). Highly significant differences (Duncan's test) between the control and each of the ZP groups (p<0.05), as well as between the vehicle and the ZP groups (P<0.001) was shown for both the absolute and relative infarct values. In addition, ZnDDC at 3x7.6 μg/kg also significantly decreased both the absolute and relative infarct volumes in this model (Figure 17A) .
Clear evidence of protection by ZP was also observed with the neurological score (Figure 18) . In the vehicle treated groups 4 out of the 10 mice (40%) showed no apparent neurological deficits (zero neurological score) even though they had significant infarctions. Such animals were not common in the control group (12.5%) . In contrast, in the ZP- treated groups the number of mice developing infarct but having no neurological deficit increased to 84.6% in the 3x1.2 μg/kg group, 61.5 % in the 3x6 μg/kg group, and 63.6% in the 3x30 μg/kg group.
Moreover, no severe deficits (neurological score of 3 or 4) were observed in the ZP- treated groups (the only exception was one mouse with the score of 3 in the 3x30 μg/kg group) . One-way ANOVA showed a statistically significant dependence of neurological score on ZP (F=2.9, P<0.05). The difference between the vehicle and the 3x1.2 μg/kg group was significant (P<0.01, post hoc Duncan's test), while the difference between the vehicle and the two other ZP doses tended to be significant (P<0.06, trend) (Figure 20).
TABLE 2. Neurological deficit in 4VO rats (neurological score, mean±S.E.M.)
Figure imgf000057_0001
ZP administered 3, 4, and 6 h after the ischemic episode.
TABLE 3. Infarct volumes and neurological deficit in mice following 60 min MCAO (mean±S.E.M.)
Figure imgf000057_0002
P<0.06 vs. vehicle- treated group;
**, P<0.01 vs. vehicle -treated group;
***, P<0.001 vs. vehicle- treated group.
\ P<0.05 vs. control group; **, P<0.01 vs. control group EXAMPLE 7 In vivo heart model - ischemic injury in pigs.
Experimental model: Adult male pigs weighing 2712 kg will be used for this study. The pigs will be premedicated with subcutaneous diazepam 5 mg and azaperon intramuscularly 5 mg/kg. Anesthesia will be initiated with intravenous thiopental 300-500 mg, and will be maintained with intravenous thiopental. After intubation, the pigs will be mechanically ventilated with a Drager respirator AV-1 (02 in room air: Fi02, 0.3; PC02, controlled) , and central venous and arterial lines will be introduced. Before a midline thoracotomy, a 7.5-mg bolus of the analgeticum piritramid will be given intravenously. The pericardium will be opened and fixed to the border of the sternum. A left atrial pressure (LAP) catheter will be introduced. Coronary occlusion by a snare will be applied by tunneling the left anterior descending (LAD) coronary artery with a monofil suture between the proximal and medial third behind the first diagonal branch. At the same level, the vena cordis magna will be cannulated for blood analysis with a small catheter. A myocardial P02 probe will then be implanted into the expected center of the area at risk. A temperature probe will be positioned next to the P02 sensor. Baseline values will be acquired during a 1-hour preoperative period. Coronary occlusion will be achieved by tightening the snare around the LAD for 60 min. The snare will then be loosened to initiate reperfusion. Two models will be tested: I. An acute non- survival model with a total of 6 h reperfusion, and II. A recovery model with 7 days of reperfusion. Each of the two models will consist of 5 groups: Sham operated (Sham), Ischemia/reperfusion alone (I/R) , I/R plus vehicle alone (I/R + DMSO) , I/R plus zinc pyrithione (I/R + ZP) , and I/R plus Zinc-diethyldithiocarbamate (I/R + ZD) . The zinc ionophores will be infused into the LAD in three boluses of 5ml saline, at 0, 1, and 2h after the initiation of reperfusion to give a cumulative dose of 1 μg/kg body weight. Vehicle will be administered in a similar and blinded fasion. Hemodynamic and P02 measurements and blood samples will be obtained at 5 , 10, 20, 30, and 60 minutes after coronary occlusion; and after 5, 10, 20, 30, 60, 90, and 120 minutes of coronary reperfusion. Global and regional contractility will be recorded by regional 2Dultrasound before, after 15 and 60 minutes of coronary occlusion, and after 15, 60, and 120 minutes of reperfusion. At the end of reperfusion, the pigs will be killed, and their hearts will be recovered for further analysis .
ECG, right atrial, pulmonary artery, and arterial pressure and LAP will be recorded on a Siemens Sirecust 404-1 at different time points. Cardiac output will be determined by thermodilution (5 mL NaCl 0.9%, room temperature) and by continuous measurement with a Baxter Vigilance monitor. The Licox catheter probe measurement system will be used in which the flexible
Licox catheter P02 microprobe is in direct contact with the myocardium. One milliliter of heparinized venous and arterial blood will be drawn with polypropylene syringes from the femoral artery and the vena cordis magna. Lactate and arteriovenous 02 differences will be determined before, after 60 minutes of coronary occlusion, and after 10 and 120 minutes of reperfusion. Blood gas analysis will be performed with the Radiometer Copenhagen Arterial Bloodgas Laboratory 3. For lactate determination, samples will be centrifuged at 2000g (10 minutes at 4°C) , plasma will be decanted, and lactate will be measured with the Lactate Analyzer model 23L from Yellow Springs Instrument Co Inc. Venous blood samples (3 ml) will be collected in polypropylene tubes containing citrate and will be centrifuged at 2000g for 15 minutes at 4°C. Plasma creatine kinase activity will be determined and expressed as international units per milliliter. Troponin-T will be measured.
With model I (non- survival) , after 6 h of reperfusion, the LAD will be reoccluded. Then, 40 ml of Evan's blue (2% wt/vol solution) will be injected into the pulmonary artery to stain perfused myocardium.
Unstained myocardium will be defined as the area at risk. After cardioplegia with 20 mL potassium chloride IV (20%) , the heart will be excised. The right ventricle, the large vessels, and fat tissue will be removed. The left ventricle will then be sliced perpendicular to the axis of the left side of the heart from the apex to the AV groove in 4 -mm slices. The unstained part of the left ventricular myocardium will be separated from the Evan's blue-stained portion and immersed in a 0.09-mol/L sodium phosphate buffer, pH 7.4, containing 1% triphenyltetrazolium chloride (TTC, Sigma) and 8% dextran (molecular weight, 77.800) for 20 minutes at 37°C. The TTC dye will form a dark-red formazan complex in the presence of viable myocardial cells that contain active dehydrogenases and cofactors. Dead cells will remain unstained. The ischemic but non-necrotic, red- stained tissue will be separated from the unstained, infarcted tissue. The three tissue sections—nonischemic (area not at risk) , ischemic non-necrotic, and ischemic necrotic tissue will be weighed. The arterial pressure-rate product (mean arterial pressure times heart rate) will be taken as a global parameter of myocardial contractility. TUNEL staining (Fliss et al . (1996) supra) , as described above, will be used to identify the percent of apoptotic myocytes in the myocardial tissue.

Claims

WHAT IS CLAIMED IS:
1. A method of treating apoptosis comprising administering to a patient in need thereof a pharmaceutically effective amount of a zinc ionophore and a pharmaceutically acceptable carrier.
2. The method of claim 1, wherein the zinc ionophore is selected from the group consisting of zinc pyrithione, heterocyclic amines, dithiocarbamates and Vitamins .
3. The method of claim 2, wherein the zinc ionophore is zinc pyrithione.
4. The method of claim 2, wherein said heterocyclic amine is selected from the group consisting of 5 , 7 -Diiodo- 8 -hydroxyquinoline and 8 -Hydroxyquinoline .
5. The method of claim 2, wherein said dithiocarbamate is selected from the group consisting of pyrrolidine dithiocarbamate, zinc-diethyldithiocarbamate, disulfiram and zinc-dimethyldithiocarbamate.
6. The method of claim 2, wherein said vitamin is selected from the group consisting of Vitamin E and Vitamin A.
7. The method of claim 1, wherein the effective amount of a zinc ionophore ranges from about 0.005 μg per kg of body weight to about 2.0 mg per kg of body weight.
8. The method of claim 7, wherein the effective amount of a zinc ionophore ranges from about 0.2μg per kg of body weight to about 600μg per kg of body weight.
9. The method of claim 1, wherein the zinc ionophore is administered intravenously, intramuscularly, subcutaneously, intracerebroventricularly, orally or topically.
10. A method of protecting against the harmful effects of injurious agents selected from the group consisting of oxidants, TNFα, neurotoxins, and radiation comprising administering to a patient in need of such protection an effective amount of a zinc ionophore and a pharmaceutically acceptable carrier.
11. The method of claim 10, wherein the zinc ionophore is selected from the group consisting of zinc pyrithione, heterocyclic amines, dithiocarbamates and Vitamins .
12. The method of claim 11, wherein the zinc ionophore is zinc pyrithione.
13. The method of claim 11, wherein said heterocyclic amine is selected from the group consisting of 5 , 7 -Diiodo- 8 -hydroxyquinoline and 8 -Hydroxyquinoline.
14. The method of claim 11, wherein said dithiocarbamate is selected from the group consisting of pyrrolidine dithiocarbamate, zinc-diethyldithiocarbamate, disulfiram and zinc-dimethyldithiocarbamate.
15. The method of claim 11, wherein said vitamin is selected from the group consisting of Vitamin E and Vitamin A.
16. The method of claim 10, wherein the effective amount of a zinc ionophore ranges from about
0.005 μg per kg of body weight to about 2.0 mg per kg of body weight.
17. The method of claim 16, wherein the effective amount of a zinc ionophore ranges from about 0.2μg per kg of body weight to about 600μg per kg of body weight.
18. The method of claim 10, wherein the zinc ionophore is administered intravenously, intramuscularly, subcutaneously, intracerebroventricularly, orally or topically.
19. A pharmaceutical composition comprising a zinc ionophore and a pharmaceutically acceptable carrier.
20. A method of treating ischemia comprising administering to a patient in need of such protection an effective amount of a zinc ionophore and a pharmaceutically acceptable carrier.
21. The method of claim 20, wherein the zinc ionophore is selected from the group consisting of zinc pyrithione, heterocyclic amines, dithiocarbamates and Vitamins .
22. The method of claim 21, wherein the zinc ionophore is zinc pyrithione.
23. The method of claim 21, wherein said heterocyclic amine is selected from the group consisting of 5, 7 -Diiodo-8 -hydroxyquinoline and 8 -Hydroxyquinoline.
24. The method of claim 21, wherein said dithiocarbamate is selected from the group consisting of pyrrolidine dithiocarbamate, zinc-diethyldithiocarbamate, disulfiram and zinc-dimethyldithiocarbamate.
25. The method of claim 21, wherein said vitamin is selected from the group consisting of Vitamin E and Vitamin A.
26. The method of claim 20, wherein the effective amount of a zinc ionophore ranges from about 0.005 μg per kg of body weight to about 2.0 mg per kg of body weight.
27. The method of claim 26, wherein the effective amount of a zinc ionophore ranges from about 0.2μg per kg of body weight to about 600μg per kg of body weight.
PCT/IB2000/001005 1999-06-23 2000-06-23 Zinc ionophores as anti-apoptotic agents WO2001000193A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2001505903A JP2003503346A (en) 1999-06-23 2000-06-23 Zinc ionophore as an anti-apoptotic agent
AU18396/01A AU1839601A (en) 1999-06-23 2000-06-23 Zinc ionophores as anti-apoptotic agents
EP00979231A EP1189604A2 (en) 1999-06-23 2000-06-23 Zinc ionophores as anti-apoptotic agents
CA002375852A CA2375852A1 (en) 1999-06-23 2000-06-23 Zinc ionophores as anti-apoptotic agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US14063299P 1999-06-23 1999-06-23
US60/140,632 1999-06-23

Publications (2)

Publication Number Publication Date
WO2001000193A2 true WO2001000193A2 (en) 2001-01-04
WO2001000193A3 WO2001000193A3 (en) 2001-12-06

Family

ID=22492127

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2000/001005 WO2001000193A2 (en) 1999-06-23 2000-06-23 Zinc ionophores as anti-apoptotic agents

Country Status (5)

Country Link
EP (1) EP1189604A2 (en)
JP (1) JP2003503346A (en)
AU (1) AU1839601A (en)
CA (1) CA2375852A1 (en)
WO (1) WO2001000193A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002080943A1 (en) * 2001-04-04 2002-10-17 Henry Fliss Zinc ionophores as anti-stress agents
WO2002089919A2 (en) * 2001-05-08 2002-11-14 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Usage of zinc for preventing tnf-induced pathology during cancer therapy
JP2006515211A (en) * 2003-01-15 2006-05-25 ストライカー コーポレイション ア コーポレイション オブ ザ ステート オブ ミシガン Emergency bed loading and unloading equipment
US10369108B2 (en) 2013-03-15 2019-08-06 Mylan Laboratories, Inc. Hot melt granulation formulations of poorly water-soluble active agents
EP3801567A4 (en) * 2018-05-25 2022-04-20 Patel, Oneel Methods and compositions for inhibiting a noxious insult

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006206538A (en) * 2005-01-31 2006-08-10 Institute Of Physical & Chemical Research Functional controlling agent for antigen presenting cell
WO2021035060A1 (en) * 2019-08-21 2021-02-25 Brain Chemistry Labs Compositions comprising a metal and l-serine, and uses thereof

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 1 May 1999 (1999-05-01) CARMODY RUAIDHRI J ET AL: "Reactive oxygen species as mediators of photoreceptor apoptosis in vitro." Database accession no. PREV199900285409 XP002165008 & EXPERIMENTAL CELL RESEARCH, vol. 248, no. 2, 1 May 1999 (1999-05-01), pages 520-530, ISSN: 0014-4827 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 1991 ZALEWSKI P D ET AL: "PHYSIOLOGICAL ROLE FOR ZINC IN PREVENTION OF APOPTOSIS GENE-DIRECTED DEATH" Database accession no. PREV199293030435 XP002165006 & BIOCHEMISTRY INTERNATIONAL, vol. 24, no. 6, 1991, pages 1093-1102, ISSN: 0158-5231 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 1993 ZALEWSKI PETER D ET AL: "Correlation of apoptosis with change in intracellular labile Zn(II) using Zinquin ((2-methyl-8-p-toluenesulphonamido-6-quino lyloxy)acetic acid), a new specific fluorescent probe for Zn(II)." Database accession no. PREV199497071032 XP002165005 & BIOCHEMICAL JOURNAL, vol. 296, no. 2, 1993, pages 403-408, ISSN: 0264-6021 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 1994 TREVES SUSAN ET AL: "Apoptosis is dependent on intracellular zinc and independent of intracellular calcium in lymphocytes." Database accession no. PREV199497270268 XP002165004 & EXPERIMENTAL CELL RESEARCH, vol. 211, no. 2, 1994, pages 339-343, ISSN: 0014-4827 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 1995 VERHAEGEN STEVEN ET AL: "Inhibition of apoptosis by antioxidants in the human HL-60 leukemia cell line." Database accession no. PREV199598551157 XP002165013 & BIOCHEMICAL PHARMACOLOGY, vol. 50, no. 7, 1995, pages 1021-1029, ISSN: 0006-2952 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 1996 MATHIEU JACQUES ET AL: "Radiation-induced apoptosis in thymocytes: Inhibition by diethyldithiocarbamate and zinc." Database accession no. PREV199799341723 XP002165009 & RADIATION RESEARCH, vol. 146, no. 6, 1996, pages 652-659, ISSN: 0033-7587 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 1996 MATSUSHITA KOHJI ET AL: "Effect of systemic zinc administration on delayed neuronal death in the gerbil hippocampus." Database accession no. PREV199799382655 XP002165007 & BRAIN RESEARCH, vol. 743, no. 1-2, 1996, pages 362-365, ISSN: 0006-8993 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; January 1998 (1998-01) TAGAMI M ET AL: "Vitamin E prevents apoptosis in hippocampal neurons caused by cerebral ischemia and reperfusion in stroke-prone spontaneously hypertensive rats." Database accession no. PREV199800147037 XP002165012 & STROKE, vol. 29, no. 1, January 1998 (1998-01), page 279 23rd International Joint Conference on Stroke and Cerebral Circulation;Orlando, Florida, USA; February 5-7, 1998 ISSN: 0039-2499 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; May 1999 (1999-05) TAGAMI MOTOKI ET AL: "Vitamin E prevents apoptosis in hippocampal neurons caused by cerebral ischemia and reperfusion in stroke-prone spontaneously hypertensive rats." Database accession no. PREV199900294771 XP002165010 & LABORATORY INVESTIGATION, vol. 79, no. 5, May 1999 (1999-05), pages 609-615, ISSN: 0023-6837 *
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; November 1998 (1998-11) TAGAMI MOTOKI ET AL: "Vitamin E prevents apoptosis in cortical neurons during hypoxia and oxygen reperfusion." Database accession no. PREV199900046515 XP002165011 & LABORATORY INVESTIGATION, vol. 78, no. 11, November 1998 (1998-11), pages 1415-1429, ISSN: 0023-6837 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002080943A1 (en) * 2001-04-04 2002-10-17 Henry Fliss Zinc ionophores as anti-stress agents
WO2002089919A2 (en) * 2001-05-08 2002-11-14 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Usage of zinc for preventing tnf-induced pathology during cancer therapy
WO2002089919A3 (en) * 2001-05-08 2003-02-20 Vlaams Interuniv Inst Biotech Usage of zinc for preventing tnf-induced pathology during cancer therapy
JP2006515211A (en) * 2003-01-15 2006-05-25 ストライカー コーポレイション ア コーポレイション オブ ザ ステート オブ ミシガン Emergency bed loading and unloading equipment
US10369108B2 (en) 2013-03-15 2019-08-06 Mylan Laboratories, Inc. Hot melt granulation formulations of poorly water-soluble active agents
EP3801567A4 (en) * 2018-05-25 2022-04-20 Patel, Oneel Methods and compositions for inhibiting a noxious insult

Also Published As

Publication number Publication date
EP1189604A2 (en) 2002-03-27
WO2001000193A3 (en) 2001-12-06
AU1839601A (en) 2001-01-31
CA2375852A1 (en) 2001-01-04
JP2003503346A (en) 2003-01-28

Similar Documents

Publication Publication Date Title
US6495538B2 (en) Zinc ionophores as therapeutic agents
Amemiya et al. Anti-apoptotic and neuroprotective effects of edaravone following transient focal ischemia in rats
Yan et al. The protective effect and its mechanism of 3-n-butylphthalide pretreatment on cerebral ischemia reperfusion injury in rats.
US6066333A (en) Pharmaceutical control of inflammation
Rastmanesh Potential of melatonin to treat or prevent age-related macular degeneration through stimulation of telomerase activity
US20100152144A1 (en) Estrogen beta receptor agonists to prevent or reduce the severity of cardiovascular disease
US6866864B2 (en) Compositions and methods of use in the treatment of angiogenesis and vascular-related disorders
Kong et al. Melatonin induces Akt phosphorylation through melatonin receptor-and PI3K-dependent pathways in primary astrocytes
BRPI0712139A2 (en) use of cannabinoid receptor agonists as hypothermia-inducing drugs for the treatment of ischemia
Lejoyeux et al. Serotonin syndrome: incidence, symptoms and treatment
WO2014028399A1 (en) Laquinimod for treatment of cannabinoid receptor type 1 (cb1) mediated disorders
WO2017082707A1 (en) Composition for treating apoplexy through nasal administration
EP0782441A1 (en) Pharmaceutical control of inflammation
US20020183300A1 (en) Zinc ionophores as anti-stress agents
Sun et al. SIRT1 activation attenuates microglia-mediated synaptic engulfment in postoperative cognitive dysfunction
EP1189604A2 (en) Zinc ionophores as anti-apoptotic agents
US6407090B1 (en) Zinc ionophores as anti-apoptotic agents
WO2003082216A2 (en) Neuroprotectant methods, compositions, and screening methods thereof
Gu et al. Remote ischemic preconditioning protects against spinal cord ischemia–reperfusion injury in mice by activating NMDAR/AMPK/PGC-1α/SIRT3 signaling
JP2021504466A (en) Treatment of tachycardia
WO2004096222A1 (en) Use of 1-(5-isoquinolinesulfonyl)homopiperazine, its active metabolites, isomers and salts for treating and preventing hypopigmentary disorders
US11857594B2 (en) Curcumin compositions and methods of use as an NK3 antagonist
McCabe et al. Inhibition of guinea-pig platelet function in vivo and ex vivo using the thromboxane A2 antagonists, AH23848 and GR32191
RU2692063C2 (en) Medicinal agent for preventing and treating cerebral diseases and method of treating cerebral diseases
KR20220107430A (en) Composition for preventing or treating parkinson&#39;s disease comprising evernic acid

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

ENP Entry into the national phase in:

Ref document number: 2375852

Country of ref document: CA

Ref country code: CA

Ref document number: 2375852

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase in:

Ref country code: JP

Ref document number: 2001 505903

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 2000979231

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 18396/01

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2000979231

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: 2000979231

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2000979231

Country of ref document: EP