US20120237449A1 - Methods for treating neural cell swelling - Google Patents

Methods for treating neural cell swelling Download PDF

Info

Publication number
US20120237449A1
US20120237449A1 US13/483,824 US201213483824A US2012237449A1 US 20120237449 A1 US20120237449 A1 US 20120237449A1 US 201213483824 A US201213483824 A US 201213483824A US 2012237449 A1 US2012237449 A1 US 2012237449A1
Authority
US
United States
Prior art keywords
channel
atp
swelling
compound
test compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/483,824
Inventor
J. Marc Simard
Mingkui Chen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Veterans Affairs VA
Original Assignee
University of Maryland at Baltimore
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Maryland at Baltimore filed Critical University of Maryland at Baltimore
Priority to US13/483,824 priority Critical patent/US20120237449A1/en
Assigned to THE UNITED STATES OF AMERICAN AS REPRESENTED BY THE DEPT. OF VETERANS AFFAIRS reassignment THE UNITED STATES OF AMERICAN AS REPRESENTED BY THE DEPT. OF VETERANS AFFAIRS ASSIGNMENT OF JOINT UNDIVIDED RIGHT, TITLE AND INTEREST Assignors: UNIVERSITY OF MARYLAND, BALTIMORE
Assigned to THE UNITED STATES OF AMERICA AS REPRESENTED BY THE DEPT. OF VETERANS AFFAIRS reassignment THE UNITED STATES OF AMERICA AS REPRESENTED BY THE DEPT. OF VETERANS AFFAIRS CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNEE NAME SHOULD READ: THE UNITED STATES OF AMERICA AS REPRESENTED BY THE DEPT. OF VETERANS AFFAIRS PREVIOUSLY RECORDED ON REEL 028306 FRAME 0937. ASSIGNOR(S) HEREBY CONFIRMS THE THE UNITED STATE OF AMERICAN AS REPRESENTED BY THE DEPT. OF VETERANS AFFAIRS. Assignors: UNIVERSITY OF MARYLAND, BALTIMORE
Publication of US20120237449A1 publication Critical patent/US20120237449A1/en
Assigned to UNIVERSITY OF MARYLAND, BALTIMORE reassignment UNIVERSITY OF MARYLAND, BALTIMORE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SIMARD, J. MARC, CHEN, MINGKUI
Priority to US14/184,947 priority patent/US9107932B2/en
Priority to US14/815,154 priority patent/US20150338393A1/en
Priority to US15/644,450 priority patent/US10533988B2/en
Priority to US15/895,945 priority patent/US20180172671A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5058Neurological cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/17Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine
    • A61K31/175Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine having the group, >N—C(O)—N=N— or, e.g. carbonohydrazides, carbazones, semicarbazides, semicarbazones; Thioanalogues thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • 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
    • A61K31/4015Heterocyclic 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 having oxo groups directly attached to the heterocyclic ring, e.g. piracetam, ethosuximide
    • 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
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • 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
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4453Non condensed piperidines, e.g. piperocaine only substituted in position 1, e.g. propipocaine, diperodon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/566Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol having an oxo group in position 17, e.g. estrone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/64Sulfonylureas, e.g. glibenclamide, tolbutamide, chlorpropamide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • A61K49/0008Screening agents using (non-human) animal models or transgenic animal models or chimeric hosts, e.g. Alzheimer disease animal model, transgenic model for heart failure
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics
    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J19/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J19/0093Microreactors, e.g. miniaturised or microfabricated reactors
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B32LAYERED PRODUCTS
    • B32BLAYERED PRODUCTS, i.e. PRODUCTS BUILT-UP OF STRATA OF FLAT OR NON-FLAT, e.g. CELLULAR OR HONEYCOMB, FORM
    • B32B37/00Methods or apparatus for laminating, e.g. by curing or by ultrasonic bonding
    • B32B37/12Methods or apparatus for laminating, e.g. by curing or by ultrasonic bonding characterised by using adhesives
    • B32B37/1284Application of adhesive
    • B32B37/1292Application of adhesive selectively, e.g. in stripes, in patterns
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0622Glial cells, e.g. astrocytes, oligodendrocytes; Schwann cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5076Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving cell organelles, e.g. Golgi complex, endoplasmic reticulum
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B29WORKING OF PLASTICS; WORKING OF SUBSTANCES IN A PLASTIC STATE IN GENERAL
    • B29LINDEXING SCHEME ASSOCIATED WITH SUBCLASS B29C, RELATING TO PARTICULAR ARTICLES
    • B29L2031/00Other particular articles
    • B29L2031/756Microarticles, nanoarticles
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the present invention relates to a novel ion channel found in neural cells which participates in the cation flux involved in cell swelling.
  • the invention also provides a method of screening for compounds that inhibit the activity of the ion channel. Methods to screen for and identify antagonists of the NC Ca-ATP channel are provided.
  • the invention further provides therapeutic methods for using compounds and compositions that inhibit the ion channel activity to inhibit or prevent the swelling of neural cells in brain. It has been discovered that neural cell swelling is mediated by the opening of a novel non-selective monovalent cationic ATP sensitive channel (the NC Ca-ATP channel) and that this channel is coupled to sulfonylurea receptor type 1.
  • neural cell swelling and cell death can be inhibited by blocking the NC Ca-ATP channel of the present invention, particularly by antagonizing receptors coupled to this channel, such as antagonizing the SUR1.
  • the invention also encompasses the use of such compounds and compositions, that modulate NC Ca-ATP channel activity to treat brain swelling.
  • the present invention relates to methods for the treatment of brain swelling that results from brain trauma or cerebral ischemia, due to neural cell swelling and cell death.
  • the normal response of the surrounding brain is to mount a cellular response that includes formation of reactive astrocytes that are believed to be important to “contain” and “clean-up” the injury site.
  • Swelling of neural cells is part of the cytotoxic or cell swelling response that characterizes brain damage in cerebral ischemia and traumatic brain injury, and is a major cause of morbidity and mortality. See, Staub et al., 1993; Kimelberg et al., 1995.
  • a number of mediators have been identified that initiate swelling of neural cells, including elevation of extracellular K + , acidosis, release of neurotransmitters and free fatty acids.
  • Cytotoxic edema is a well-recognized phenomenon clinically that causes brain swelling, which worsens outcome and increases morbidity and mortality in brain injury and stroke.
  • apoptotic death of reactive astrocytes have been studied. See, Tanaka et al., 2000; Yu et al., 2001. The mechanisms responsible for necrotic cell death have not been characterized. Apoptotic cell death is preceded by cell shrinkage and net loss of K. See, Yu et al., 1997; Yu et al., 1999. By contrast, in necrotic cell death, the plasma membrane is ruptured, causing cytosolic contents to be released and thereby triggering tissue inflammation. See, Leist and Nicotera, 1997. Necrotic cell death may be more deleterious to nearby viable tissues, given the secondary inflammatory damage that is initiated.
  • Necrotic cell death is initiated by osmotic swelling following influx of Na + , the major extracellular osmolyte.
  • accumulation of Na + intracellularly is regarded as a passive process that does not require activation of specific effectors but that is due instead to defective outward Na′ pumping under conditions of low [ATP]i.
  • [ATP]i the major extracellular osmolyte.
  • Cell blebbing or swelling an indication of intracellular Na′ overload, is generally regarded as an early sign of necrotic cell death. See, Leist and Nicotera, 1997; Majno and Joris, 1995.
  • Non-selective cation channel One subtype of ATP sensitive cation channel is the non-selective cation channel, which are channels that are sensitive to Ca2 + and ATP. More specifically, non-selective cation channels are activated by intracellular Ca2 + ([Ca2 + ] I and inhibited by intracellular ATP ([ATP] i ). Although Ca2 + and ATP sensitive cation channels had been identified in a number of non-neural cell types, they have not been identified in astrocytes or any other neural cells.
  • non-astrocyte channels comprise a heterogeneous group with incompletely defined characteristics. They exhibit single-channel conductances in the range of 25-35 pS, discriminate poorly between Na + and K + , are impermeable to anions, for the most part impermeable divalent cations, and they are blocked by similar concentrations of the adenine nucleotides ATP, ADP and AMP on the cytoplasmic side. The function of these non-selective ATP sensitive cation channels in these non-neural cell types remains enigmatic, in part because unphysiological concentrations of Ca2 + are generally required for channel activation.
  • KAP channels ATP-sensitive potassium channel
  • ATP-sensitive potassium channel KAP channels
  • SUR high affinity sulfonylurea receptor
  • SUR1 Several isoforms of the SUR, termed SUR1, SUWA, S W B, and SUR2c, have been identified and cloned. See, Aguilar-Bryan et. al., 1995; Inagaki et. al., 1996; Isomoto et. al., 1996; Lawson, 2000. These receptors belong to the ATP-binding cassette (ABC) transporter family, of which the cystic fibrosis transmembrane conductance regulator (CFTR), another ion channel modulator, is also a member. See, Higgins, 1992; Aguilar-Bryan et. al., 1995. Notably, the CFTR has major therapeutic importance, since its genetic absence causes cystic fibrosis, a fatal disease.
  • ABSC ATP-binding cassette
  • the sulfonylurea receptor imparts sensitivity to antidiabetic sulfonylureas such as glibenclamide and tolbutamide.
  • SUR is responsible for activation of the potassium channel by a chemically diverse group of agents termed K + channel openers (SUR-activators), such as diazoxide, pinacidil, and cromakalin.
  • K + channel openers such as diazoxide, pinacidil, and cromakalin.
  • K ATP channel in pancreatic ⁇ cells is formed from SUR1 linked with a K+ channel
  • cardiac and smooth muscle K ATP channels are formed from SUR2A and SUR2B, respectively, linked to K+ channels. See, Fujita ad Kurachi, 2000.
  • the present invention is directed to a newly characterized non-selective calcium and ATP sensitive monovalent cation channel, termed the NC Ca-ATP channel, which is present in neural cells and linked to an SUR.
  • the present invention further provides a method to screen for or identify antagonists to NC Ca-ATP channel activity.
  • the present invention provides a method for the therapeutic use of antagonists, such as sulfonylureas and other SUR1 blockers, to inhibit this channel's activity and thereby prevent neural cell swelling and cell death and the concomitant nervous system damage that includes brain swelling and brain damage.
  • antagonists such as sulfonylureas and other SUR1 blockers
  • the invention is based, in part, on the discovery of a specific channel, the NC Ca-ATP channel, which is expressed in reactive neural cells after brain trauma.
  • the present invention is directed to purified compositions containing a novel Ca 2+ -activated, [ATP] i , sensitive nonspecific cation channel, hereinafter the NC Ca-ATP channel.
  • the compositions comprise mammalian neural cells or membrane preparations expressing the NC Ca-ATP channel, most preferably the mammalian neural cells are freshly isolated reactive astrocytes.
  • a preferred example of such a purified composition containing the NC Ca-ATP channel is a membrane preparation derived from native reactive astrocytes.
  • the NC Ca-ATP channel opens and the cells swell and die. However, if the NC Ca-ATP channel is blocked on such cells, the cells do not swell and die.
  • the invention is also based, in part, on the discovery that the NC Ca-ATP channel is regulated by a type 1 sulfonylurea receptor, and that antagonists of this receptor are capable of blocking the NC Ca-ATP channel and inhibit neural cell swelling.
  • the NC Ca-ATP channel of the present invention is distinguished by certain functional characteristics, the combination of which distinguishes it from known ion channels.
  • the characteristics that distinguish the NC Ca-ATP channel of the present invention include, but are not necessarily limited to, the following: 1) it is a non-selective cation channels that readily allows passage of Na, K and other monovalent cations; 2) it is activated by an increase in intracellular calcium, and/or by a decrease in intracellular ATP; 3) it is regulated by sulfanylurea receptor type 1 (SURD, which heretofore had been considered to be associated exclusively with K ATP channels such as those found in pancreatic ⁇ cells.
  • SURD sulfanylurea receptor type 1
  • the NC Ca-ATP channel of the present invention has a single-channel conductance to potassium ion (K + ) between 20 and 50 pS.
  • the NC Ca-ATP channel is also stimulated by Ca 2+ on the cytoplasmic side of the cell membrane in a physiological concentration range, where said concentration range is from 10 ⁇ 8 to 10 ⁇ 5 M.
  • the NC Ca-ATP channel is also inhibited by cytoplasmic ATP in a non-physiological concentration range, where said concentration range is from 10 ⁇ 1 to 10 M.
  • the NC Ca-ATP channel is also permeable to the following cations; K + , Cs + , Li + , Na + ; to the extent that the permeability ratio between any two of said cations is greater than 0.5 and less than 2.
  • the invention relates to assays designed to screen for compounds or compositions that modulate the NC Ca-ATP channel, particularly compounds or compositions that act as antagonists of the channel, and thereby modulate neural cell swelling and the concomitant brain swelling.
  • cell-based assays or non-cell based assays can be used to detect compounds that interact with, e.g., bind to, the outside (i.e., extracellular domain) of the NC Ca-ATP channel and/or its associated SUR1.
  • the cell-based assays have the advantage in that they can be used to identify compounds that affect NC Ca-ATP channel biological activity (i.e., depolarization).
  • the invention also provides a method of screening for and identifying antagonists of the NC Ca-ATP channel, by contacting neural cells with a test compound and determining whether the test compound inhibits the activity of the NC Ca-ATP channel.
  • methods for identifying compounds that are antagonists of the NC Ca-ATP are provided.
  • therapeutic compounds of the present invention, including NC Ca-ATP antagonists are identified by the compound's ability to block the open channel or to prevent channel opening, by quantifying channel function using electrophysiological techniques to measure membrane current through the channel.
  • NC Ca-ATP antagonists include compounds that are NC Ca-ATP channel inhibitors, NC Ca-ATP channel blockers, SUR1 antagonists, SUR1 inhibitors, and/or a compounds that reduce the magnitude of membrane current through the channel.
  • channel function can be measured in a preparation of neural cells from a human or animal, and the test compound can be brought into contact with the cell preparation by washing it over the cell preparation in solution.
  • the invention further provides a method of screening for sulfonylurea compounds that may act as antagonists of the NC Ca-ATP channel.
  • the present invention relates to drug screening assays to identify compounds for the treatment of brain swelling, such as the swelling that occurs after brain injury or cerebral ischemia by using the NC Ca-ATP channel as a target.
  • the invention also relates to compounds that modulate neural cell swelling via the NC Ca-ATP channel.
  • the present invention also relates to the treatment of brain swelling by targeting the NC Ca-ATP channel.
  • the invention also encompasses agonists and antagonists of the NC Ca-ATP channel, including small molecules, large molecules, and antibodies, as well as nucleotide sequences that can be used to inhibit NC Ca-ATP channel gene expression (e.g., antisense and ribozyme molecules).
  • An antagonists of the NC Ca-ATP channel includes compounds capable of (1) blocking the channel, (2) preventing channel opening, and/or (3) reducing the magnitude of membrane current through the channel.
  • the invention also encompasses the use of such compounds and compositions, that modulate NC Ca-ATP channel activity to treat brain swelling. Further provided is a method of preventing brain swelling and the resulting brain damage through the therapeutic use of antagonists to the NC Ca-ATP channel.
  • the therapeutic antagonist can be administered to or into the brain. Such administration to the brain includes injection directly into the brain, particularly in the case where the brain has been rendered accessible to injection due to trauma to the skull.
  • the invention further provides the therapeutic use of sulfonylurea compounds as antagonists to the NC Ca-ATP channel to prevent cell swelling in brain.
  • the sulfonylurea compound is glibenclamide.
  • the sulfonylurea compound is tolbutamide.
  • FIG. 1 (comprised of FIGS. 1A , 1 B, 1 C, 1 D, 1 E and 1 F);
  • FIG. 1A shows whole cell current clamp recording before and after exposure to ouabain and before and after exposure to N a N 3 .
  • FIG. 1B shows whole cell voltage-clamp recordings during ramp pulses (a) before and (b) after exposure to N a N 3 ; (c) is the difference current.
  • FIG. 1C shows whole cell voltage-clamp recordings during step pulses (a) before and (b) after exposure to N a N 3 ; (c) is the difference current.
  • FIG. 1A shows whole cell current clamp recording before and after exposure to ouabain and before and after exposure to N a N 3 .
  • FIG. 1B shows whole cell voltage-clamp recordings during ramp pulses (a) before and (b) after exposure to N a N 3 ; (c) is the difference current.
  • FIG. 1C shows whole cell voltage-clamp recordings during step pulses (a) before and (b)
  • FIG. 1D shows cell-attached patch recording of single ion channel openings induced by N a N 3 at membrane potentials of (3) ⁇ 80 mV and (4) 80 mV, compared to control patches at membrane potentials of (1) 80 mV and (2) ⁇ 80 mV.
  • FIG. 1E shows the cell-attached patch currents of FIG. 1D , shown at higher time resolution.
  • FIG. 1F shows the cell-attached patch single-channel current-voltage relationship.
  • FIG. 2 (comprised of FIGS. 2A and 2B ): FIG. 2A shows single channel currents recorded in an inside-out patch at different membrane potentials; dotted line indicates channel closing. FIG. 2B is a plot of inside-out patch single channel amplitude vs. membrane potentials.
  • FIG. 3 (comprised of FIGS. 3A , 3 B, 3 C and 3 D);
  • FIG. 3A shows single channel currents recorded in an inside-out patch with various alkaline ions substituting for K + in the pipette; dotted line indicates channel closing.
  • FIG. 3B is a plot of channel amplitude vs. membrane potential with various alkaline ions substituting for K + in the pipette.
  • FIG. 3C is a plot of channel amplitude measured in inside-out patches vs. voltage with Ca2 + and Mg2 + substituting for K + in the pipette. To estimate channel pore size, FIG.
  • 3D is a plot illustrating the relationship between the permeability (relative to Cs + ) and the molecular radius of a series of monovalent organic cations, which included: (a) methanolmine, (b) guanidium, (c) ethanolamine, (d) diethylamine, (e) piperazine, (f) Tris, and (g) N-methylglucamine, data indicating an equivalent pore size of 0.67 nm.
  • FIG. 4 (comprised of FIGS. 4A and 4B );
  • FIG. 4A shows single channel recordings in an inside-out patch in the absence and presence of cytoplasmic ATP.
  • FIG. 4B is a plot of normalized open channel probability (n ⁇ Po) vs. concentration of cytoplasmic ATP.
  • FIG. 5 (comprised of FIGS. 5A and 5B );
  • FIG. 5A shows current records from an inside-out patch exposed to different concentrations of [Ca2 + ] i .
  • FIG. 5B the values of n ⁇ Po measured at the membrane potentials and [Ca2 + ] i indicated.
  • FIG. 6 is a plot of mean single channel amplitudes obtained in an inside-out patch configuration at different potentials studied and with different [Mg 2+ ] i ; the dotted line indicates 35 pS conductance.
  • FIG. 7 shows that presence of SUR1 mRNA and absences of Kir6.1 and Kir 6.2 in reactive astrocytes.
  • Lanes 3 and 5 in FIG. 7A show the presence of SUR1 in insulinoma RIN-m5f cells and NRAs, respectively.
  • Lanes 4 and 6 in FIG. 7A show that SUFU is absent in both cell types.
  • Lanes 3 and 4 in FIG. 7B show that Kir6.1 is present in insulinoma RIN-m5f cells and Kir6.2 is absent from the insulinoma cells, respectively.
  • Lanes 5 and 6 in FIG. 7B show that neither Kir6.1 nor Kir6.2 is present in NRAs, respectively.
  • FIG. 8 shows current recordings in an inside-out patch to illustrate the effects of tryptic digestion on channel sensitivity to glibenclamide and ATP.
  • FIG. 9 shows that the channel activator diazoxide can elicit channel activities under outside-out patch recording configuration.
  • FIG. 9A shows the outside-out patch recordings with Na azide and diazoxide applied to the extracellular side of the membrane.
  • FIG. 9B shows the current records obtained from the segments marked with the corresponding numbers in FIG. 9A , at higher temporal resolution.
  • FIG. 10 (comprised of FIGS. 10A , 10 B and 10 C)
  • FIG. 10A shows outside-out patch recordings (a) before, (b) during, and (c) after application of glibenclamide to the extracellular side of the membrane.
  • FIG. 10B shows the current records of FIG. 10A at higher temporal resolution.
  • FIG. 10C show a plot of mean single channel amplitudes at the different potentials studied; the slope of the data indicates 35 pS conductance of the glibenclamide-sensitive channel.
  • FIG. 11 shows that sulfonylurea compounds inhibit channel activities.
  • FIG. 11A shows the outside-out patch recordings with various concentrations of tolbutamide applied to the extracellular side of the membrane.
  • FIG. 11 B shows the dose-response curves for inhibition of open channel probability by glibenclamide and tolbutamide to provide a normalized open channel probability (n ⁇ Po); data were fit to a standard logistic equation, with a Hill coefficient of 1 and half-maximum inhibition of 48 nM and 16.1 ⁇ M; values plotted are means ( ⁇ SE) from 3 and 5 patches for Glibenclamide and Tolbutamide, respectively.
  • FIG. 12 (comprised of FIGS. 12A , 12 B, 12 C, 12 D, 12 E, 12 F, 12 G, 12 H and 12 I);
  • FIGS. 12A , 12 B and 12 C show the probability of channel opening in the presence of 0 ⁇ M, 3 ⁇ M, and 30 ⁇ M tolbutamide, respectively.
  • FIGS. 12D , 12 E and 12 F show the distribution of open channel dwell times in the presence of 0 ⁇ M, 3 ⁇ M, and 30 ⁇ M tolbutamide, respectively.
  • FIGS. 12G , 12 H and 12 I show the distribution of closed channel dwell times in the presence of 0 ⁇ M, 3 ⁇ M, and 30 ⁇ M tolbutamide, respectively.
  • FIG. 13 (comprised of FIGS. 13A , 13 B and 13 C)
  • FIG. 13A shows outside-out patch recordings with diazoxide applied to the extracellular side of the membrane.
  • FIG. 13B shows current records at higher temporal resolution after application of diazoxide and at different membrane potentials.
  • FIG. 13C shows a plot of mean single channel amplitudes at the different potentials studied; the slope indicates 35 pS conductance of glibenclamide-sensitive channel.
  • FIGS. 14A , 14 B and 14 C are scanning electron micrographs of freshly isolated native reactive astrocytes.
  • FIG. 14A shows the cells when formaldehyde-glutaraldehyde fixation was initiated under control conditions;
  • FIG. 14B shows the cells fixed 5 min after exposure to 1 mM NaN3.
  • FIG. 14C shows the cells fixed 25 min after exposure to 1 mM NaN3. Bar, 12 ⁇ m.
  • FIG. 15 (comprised of FIGS. 15A , 15 B and 15 C);
  • FIG. 15A has photomicrographs of the epifluorescence images of cells exposed to different compounds and labeled with gropidium iodide (upper panel a, b and c) or annexin V (lower panel d, e and f). The compounds were: control (a & d), 1 mM Na azide (b & e), 1 mM Na azide plus 1 ⁇ M glibenclamide (c & f).
  • FIG. 15B has bar graphs showing cell-counts for propidium iodide labeling; pairwise multiple comparisons indicated a significant difference (p ⁇ 0.05) with Na azide treatment;
  • FIG. 15C has bar graphs showing cell-counts for annexin V staining; pairwise multiple comparisons indicated no significant difference with any treatment.
  • the present invention relates to a novel ion channel whose function underlies the swelling of mammalian neural cells, such as in response to ATP depletion; the use of the channel to screen for channel inhibitors, and the use of inhibitors of the channel function to prevent this cell swelling response, which characterizes brain damage in cerebral ischemia and traumatic brain injury.
  • N 3 Sodium azide
  • Reactive astrocytes are astrocytes that have been activated or stimulated in vivo, such as those associated with brain or neural injury.
  • TBI traumatic brain injury
  • reactive astrocytes are found in proximity to the injury.
  • the majority of reactive astrocytes surrounding an injury site in the brain are reactive astrocytes.
  • Type 1 reactive astrocytes comprise >80% of recoverable reactive astrocytes, whereas type 2 reactive astrocytes comprise about 5%.
  • Reactive astrocytes are normally polarized under quiescent conditions.
  • neural cells includes astrocytes.
  • reactive astrocytes means astrocytes found in brain at the site of a lesion or ischemia.
  • WRAs native reactive astrocytes
  • the term “native reactive astrocytes” or WRAs” means reactive astrocytes that are freshly isolated from brain.
  • freshly isolated refers to NRAs that have been purified from brain, particularly NRAs that were purified from about 0 to about 72 hours previously. When NRAs are referred to as being “purified from brain” the word “purified” means that the NRAs are isolated from other brain tissue and/or implanted gelatin or sponge and does not refer to a process that simply harvests a population of cells from brain without further isolation of the cells.
  • the NC Ca-ATP channel found in reactive astrocytes is present only in freshly isolated cells; the NC Ca-ATP channel is lost shortly after culturing the cells.
  • NRAs provide an in vitro model that is more similar to reactive astrocytes as they exist in vivo in the brain, than astrocytes grown in culture.
  • native and “freshly isolated” are used synonymously.
  • isolated neural cells means neural cells isolated from brain.
  • Reactive astrocytes are produced in vivo and harvested from brain according to a method system similar to that described by Perillan. See, Perillan et al., 1999; Perillan et al., 2000. Harvested cells are then isolated and not cultured; rather, the freshly isolated reactive astrocytes are studied in a native state immediately after their isolation from the brain.
  • NC Ca-ATP channel of the present invention which is newly identified in NRAs and present in >90% of membrane patches from such cells, is distinguished from previously reported non-selective calcium and ATP channels by exhibiting significantly different properties.
  • These distinguishing properties of the NC Ca-ATP of the present invention include: being activated by submicromolar [Ca] and exhibiting a different sensitivity to block by various adenine nucleotides.
  • NC Ca-ATP channel of the present invention Opening of the NC Ca-ATP channel of the present invention by ATP depletion causes profound membrane depolarization, which precedes blebbing of the cell membrane.
  • the NC Ca-ATP channel opens to allow Na + influx that leads to cell swelling.
  • This channel is regulated by sulfonylurea receptor type 1 (SUR1).
  • SUR1 sulfonylurea receptor type 1
  • the channel can be blocked by sulfonylurea, such as glibenclamide and tolbutamide; treatment with glybenclamide results in significant reduction in swelling and blebbing induced by chemical ATP depletion. This channel participates in the cation flux involved in cell swelling.
  • a method of the present invention includes the use of sulfonylurea compounds to inhibit the flow of current through the NC Ca-ATP channel and inhibit blebbing related to channel opening. Also, use of sulfonylurea compounds and other compounds that inhibit the flow of current through the NC Ca-ATP channel, thus can have a therapeutic preventative effect on cell swelling in brain.
  • the membrane preparation is derived from neural cells, such as isolated native reactive astrocytes (NRAs), preferably freshly isolated native reactive astrocytes.
  • the NC Ca-ATP channel in the composition has the following characteristics: (a) it is a 35 pS type channel; (b) it is stimulated by cytoplasmic Ca2 + ; (c) it opens when cytoplasmic ATP is less than about 0.804; and (d) it is permeable to the monovalent cations K + , Cs + , Li + and Na + and it can be blocked by antagonists of the type 1 sulfonylurea receptor.
  • the composition may contain a preparation of neural cells expressing the NC Ca-ATP channel or a membrane preparation expressing the NC Ca-ATP channel, such as a membrane preparation derived from isolated native reactive astrocytes (NRAs).
  • the effect of the compound on this channel may include: (a) blocking the NC Ca-ATP channel; (b) closing the NC Ca-ATP channel; (c) preventing the NC Ca-ATP channel from opening; and (d) reducing the magnitude of membrane current through the NC Ca-ATP channel. It is also an object of the present invention to identify a compound that is an NC Ca-ATP antagonist, including an NC Ca -ATP channel inhibitor, an NC Ca-ATP channel blocker, a SUR1 antagonist, SUR1 inhibitor, and/or a compound capable of reducing the magnitude of membrane current though the channel.
  • Yet another object of the present invention is to provide a method for identifying compounds that inhibit brain swelling, comprising: (a) contacting a test compound with a composition comprising the NC Ca-ATP channel, and (b) determining whether the test compound inhibits neural cell swelling, wherein a test compound that inhibits neural cell swelling is identified as a compound for inhibiting brain swelling.
  • a further object of the present invention provides a method for identifying compounds that inhibit neural cell swelling in an animal, comprising: (a) contacting a test compound with a composition comprising the NC Ca-ATP channel and determining whether the test compound blocks the channel, and (b) administering the test compound to an animal having a brain injury or cerebral ischemia, and determining whether the test compound that inhibits brain swelling of the treated animal, wherein test compounds that inhibit brain swelling are identified as compounds that inhibit neural cell swelling in an animal.
  • the composition preferably comprises a preparation of neural cells expressing the NC Ca-ATP channel or a membrane preparation expressing the NC Ca-ATP channel, which preferably is derived from isolated native reactive astrocytes (NRAs). It is a further object of the present invention to provide the above methods using a compound that is an antagonist of a type 1 sulfonylurea receptor, such as a sulfonylurea compound, a benzamido derivative or an imidazoline derivative.
  • a type 1 sulfonylurea receptor such as a sulfonylurea compound, a benzamido derivative or an imidazoline derivative.
  • the determining step include, but are not limited to, detecting or identifying swelling of the native reactive astrocytes, such as by microscopic observation of cell appearance (normal, blebbing, swelling); measuring channel currents; measuring membrane potential; detecting expression of annexin V; detecting expression of propidium iodide; in vitro binding assays; and combinations thereof.
  • Such administration may be delivery directly to the brain, intravenous, subcutaneous, intramuscular, intracutaneous, intragastric and oral administration.
  • Examples of such compounds include antagonist of a type 1 sulfonylurea receptor, such as sulfonylureas like glibenclamide and tolbutamide, as well as other insulin secretagogues such as repaglinide, nateglinide, meglitinide, midaglizole, LY397364, LY389382, gliclazide, glimepiride, MgADP, and combinations thereof.
  • a type 1 sulfonylurea receptor such as sulfonylureas like glibenclamide and tolbutamide
  • insulin secretagogues such as repaglinide, nateglinide, meglitinide, midaglizole, LY397364, LY389382, gliclazide, glimepiride, MgADP, and combinations thereof.
  • Reactive astrocytes are produced in vivo and harvested from adult brain in the following manner: gelatin sponges (Gelfoam®, Upjohn Co., Kalamazoo Mich.) are implanted into a stab wound in the parietal lobe of 8 week old Wistar rats as described herein. Sponge pieces are harvested at 8 days and washed three times in phosphate-buffered saline (PBS, pH 7.4) to remove adherent tissue. Depending on the number of NRAs required for a particular study, the sponge pieces may be harvested earlier or later after implantation into a stab wound, with the preferred harvest being conducted from about 2 days to about 30 days after implantation, and the most preferred range being conducted from about 2 days to about 3 days after implantation.
  • gelatin sponges (Gelfoam®, Upjohn Co., Kalamazoo Mich.) are implanted into a stab wound in the parietal lobe of 8 week old Wistar rats as described herein.
  • Sponge pieces are harvested at
  • NRAs are freshly isolated from the sponge pieces in the following manner: washed pieces are placed in an Eppendorf tube containing artificial cerebrospinal fluid (aCSF) composed of (mM): 124 mM NaCl, 5.0 mM, 1.3 mM MgCl 2 , 2.0 mM CaCl 2 , 26 mM NaHCO 3 , and 10 mM D-glucose; at pH 7.4, ⁇ 290 mOsm, wherein the aCSF contains papain 20 U/ml, trypsin inhibitor 10 mg/ml and DNase 0.01% (Worthington, Lakewood, N.J.), the entirety of which is referred to as a “digestion system.”
  • aCSF cerebrospinal fluid
  • This digestion system is transferred to an incubator (humidified 90%/10% air/C02, 37° C.) for 20 minutes, and is gently triturated every 5 minutes.
  • the cell suspension is centrifuged at 3,000 rpm for 1 minute.
  • the pelleted cells are resuspended in aCSF and stored at 4° C. until studied.
  • the pelleted cells prior to resuspension in aCSF, can be further purified by removing red blood cells (RBCs) using density gradient centrifugation in Histopaque-1 077 (Sigma Diagnostics, St. Louis, Mo.). This further purification process can produce a population of cells in which ⁇ 1% are RBCs, as determined by phase contrast microscopy.
  • RBCs red blood cells
  • Histopaque-1 077 Sigma Diagnostics, St. Louis, Mo.
  • Membrane currents are amplified (Axopatch 200A, Axon Instruments, Foster City, Calif.) and sampled on-line at 5 kHz using a microcomputer equipped with a digitizing board (Digidata 1200A, Axon Instruments) and running Clampex software (version 8.0, Axon Instruments).
  • Membrane currents are recorded in intact cells using both the cell-attached and the nystatin-perforated whole-cell configurations, according to methods described in Horn and Marty, 1988.
  • Membrane currents are recorded in cell-free isolated membrane patches, using both the inside-out and outside-out configurations, such as those described in Hamill et al., 1981.
  • Patch clamp pipettes pulled from borosilicate glass (Kimax, Fisher Scientific, Pittsburgh, Pa.), have resistances of 6-8 M ⁇ a for single channel recordings and 2-4 M ⁇ a for experiments using the nystatin-perforated whole-cell technique.
  • the bath electrode is a Ag/AgCl pellet (Clark Electromedical, Reading, England) that is placed directly in the bath except when the bath [Cl ⁇ ] is altered, in which case an agar bridge made with 3 M KCl is used to connect to the bath.
  • clamping a cell membrane refers to holding the voltage across the cell membrane constant and measuring changes in membrane current as membrane resistance changes due to ion channel opening and closing (“voltage clamp”) or holding the current across the cell membrane constant and measuring changes in membrane voltage as membrane resistance changes due to ion channel opening and closing (“current clamp”).
  • voltage clamp ion channel opening and closing
  • current clamp current clamp
  • the “whole-cell” experimental configuration refers to a situation in which a recording pipette penetrates the cell membrane so that the pipette solution is continuous with the cytoplasm or the membrane under the pipette is perforated using nystatin, the external solution is in contact with the extracellular membrane, and current or voltage recordings represent measurements from the entire cell membrane.
  • the “cell-attached patch” experimental configuration refers to a situation in which the pipette contacts the cell so that the patch is still forming part of the intact cell membrane and channels in the patch are recorded.
  • the “outside-out patch” experimental configuration refers to a situation in which an excised patch of cell membrane is sealed to the tip of a recording pipette so that the pipette solution is in contact with the extracellular side of the membrane, the external solution is in contact with the cytoplasmic side of the membrane, and current or voltage recordings represent measurements from the excised patch of membrane.
  • the “inside-out patch” experimental configuration refers to a situation in which an excised patch of cell membrane is sealed to the tip of a recording pipette so that the pipette solution is in contact with the cytoplasmic side of the membrane, the external solution is in contact with the extracellular side of the membrane, and current or voltage recordings represent measurements from the excised patch of membrane.
  • patches includes, but is not limited to: inside-out patches, outside-out patches, an excised patch of a cell membrane, or a cell-attached patch.
  • membrane preparation includes patches as well as cell membranes isolated from mammalian cells or tissues. Isolated mammalian cell membranes are produced by methods well known in the art. One example of such a membrane preparation is a microsomal fraction purified from disrupted cells or a tissue sample by discontinuous sucrose gradient centrifugation.
  • Macroscopic membrane currents are measured during step pulses (600 ms) or during ramp pulses ( ⁇ 140 to +50 mV at 0.32 mV/ms) from a holding potential of ⁇ 67 mV.
  • a nystatin perforated patch technique is used, with a bath solution containing (mM): NaCl 130, KCl 10, CaCl 2 1, MgCl 2 1, HEPES 32.5, glucose 12.5, pH 7.4.
  • the pipette solution contains (mM): KCl 55, K 2 SO4 75, MgCl 2 8, and HEPES 10, pH 7.2.
  • Nystatin, 50 mg (Calbiochem) is dissolved in dimethylsulfoxide (DMSO), 1 ml.
  • a bath solution is used containing (mM): NaCl 130, KCl 10, CaCl 2 1, MgCl 2 1, HEPES 32.5, glucose 12.5, pH 7.4.
  • the pipette contains (mM): KCl 145, MgCl 2 1, CaCl 2 0.2, EGTA 5, HEPES 10, pH 7.28.
  • the measured osmolarity of the extracellular solution is ⁇ 300 mOsm (Precision Systems, Natick, Mass.).
  • a bath solution is used containing (mM): CsCl 145, CaCl 2 4.5, MgCl 2 1, EGTA 5, HEPES 32.5, glucose 12.5, pH 7.4.
  • the pipette contains (a): CsCl 145, MgCl 2 1, CaCl 2 0.2, EGTA 5, HEPES 10, pH 7.28.
  • Cs+ in the above solutions is replaced with equimolar K + .
  • Cs+ in the pipette is replaced by equimolar concentrations of individual test ions, except when using Ca2 + or Mg2 + , in which cases a concentration of 75 mh4 is used to facilitate seal formation (Cook et al., 1990).
  • the pipette solution contains (mM): CsCl 145, MgCl 2 1, CaCl 2 0.2, EGTA 5, HEPES 10, pH 7.28.
  • the standard bath solution contains (mM): CsCl 145, CaCl24.5, MgCl 2 1, EGTA 5, HEPES 32.5, glucose 12.5, pH 7.4.
  • Cs+ in the bath is replaced with equimolar concentrations of test cation.
  • Single-channel amplitudes used to calculate slope conductance are obtained by fitting a Gaussian function to an all-points amplitude histogram of records obtained at various potentials.
  • the all-points histogram is fit to a Gaussian function and the area under the fitted curve for the open channel is divided by the area under the fitted curve for the closed plus open channel.
  • Values of n ⁇ Po at different concentration of test agents are fit to a standard logistic equation using a least-squares method.
  • each permeability is obtained from its reversal potential (Erev) by fitting to the Goldman-Hodgkin-Katz (GHK) equation well known in the art. See Goldman 1943; Hodgkin and Katz, 1949. Current-voltage data are fit to the GHK equation, assuming that both K+ and the test ion are permeant.
  • GHK Goldman-Hodgkin-Katz
  • the Stoke-Einstein radius is then converted to the molecular radius using correction factors read off from Figure 6.1 in Robinson and Stokes, 1970.
  • a/a 0 [1 ⁇ ( r/R )] 2 ⁇ [1 ⁇ 2.104( r/R )+2.09( rR ) 3 ⁇ 0.95( r/R ) 5 ] (1)
  • a, a 0 , r, and R are the effective area of the pore, the total cross sectional area of the pore, radius of the solute, and radius of the pore, respectively.
  • junction potentials are determined with an electrometer by measuring the diffusion potential established across a dialysis membrane and are subtracted when appropriate. Holding currents are not subtracted from any of the recordings. Difference currents are obtained by simply subtracting current records before and after perfusing NaN 3 , with no other processing being employed.
  • NRAs The surfaces of freshly isolated NRAs are highly complex, exhibiting small membrane evaginations and fine processes that decorate the entire cell surface, as shown in the scanning electron micrograph in FIG. 14A .
  • Exposure of NRAs to NaN 3 (1 mM) causes changes in the surface appearance, characterized early-on by loss of complex structure and development of surface blebs ( FIG. 14B ), followed later by a grossly swollen appearance with complete loss of fine structure and formation of multiple large blebs ( FIG. 14C ). Therefore, NRAs undergo blebbing and swelling after NaN3-induced ATP depletion.
  • Phase contrast microscopy is also useful for assessing this process, although fine structure cannot be resolved. Blebbing is visibly apparent 10-15 minutes after exposure to NaN 3 . Morphological changes of this sort are attributable to loss of cytoskeletal integrity, combined with action of an osmotic force that causes swelling of the cell.
  • NRAs The macroscopic currents of whole cell preparations of N u s are characterized by small inward currents at negative potentials, large outward currents at positive potentials, and a flat “plateau” region at intermediate potentials. NRAs exhibit macroscopic currents that are consistent with observations in primary cultured cells of the same origin. See, Perillan et al., 1999; Perillan et al., 2000.
  • NRAs exhibited inward currents negative to the K + equilibrium potential (E K ) are usually ⁇ 100 pA, much smaller than values reported in cultured neonatal astrocytes (Ransom and Sontheimer, 1995), but consistent with findings in astrocytes freshly isolated from injured brain (Bordey and Sontheimer, 1998; Schroder et al., 1999).
  • the large outward currents in NRAs are partially blocked by charybdotoxin (100 nM), iberiotoxin (1 00 nM) and tetraethylammonium chloride (5 mM), consistent with the presence of a large conductance Ca2 + -activated K + channel. See, Perillan et al., 1999.
  • depolarization is a primary event, not secondary to cell swelling or stretch.
  • Voltage-clamp recordings show that exposure to NaN 3 results in a net increase of inward current in NRAs.
  • Recordings obtained using both ramp ( FIG. 1B ) and step pulses ( FIG. 1 C) show significantly larger currents after NaN 3 treatment, as shown by comparing the recordings before (a) and after (b) NaN 3 treatment.
  • a plot of the “difference currents”, obtained by subtracting the current-voltage curve before drug from that after drug (line c in FIG. 1B ) indicates that the new current turned on by NaN 3 reverses near 0 mV.
  • a reversal potential near 0 mV is indicative that the NaN 3 -induced current results from a non-specific cation conductance.
  • RVD regulatory volume decrease
  • NC Ca-ATP channel is seldom observed in cell attached patches from healthy cells, but becomes evident in >90% of patches after conversion to an inside-out configuration. Also, the NC Ca-ATP channel is lost shortly after culturing reactive astrocytes.
  • the channel is further characterized using membrane patches in the inside-out configuration. Records obtained during test pulses to various potentials with equal [K + ] on both sides of the membrane are shown in FIG. 2A . Amplitude histograms are constructed of events observed at potentials from ⁇ 140 mV to +100 mV, and values (mean ⁇ SE) for 4 patches are plotted and show in FIG. 2B . Fit of the data to a linear equation indicates a slope conductance of 35 pS, with an extrapolated reversal potential (E rev ) of +0.1 mV, close to the expected K + reversal potential (E K ) of 0 mV.
  • E rev extrapolated reversal potential
  • the channel transports a variety of alkaline ions ( FIG. 3A ), indicating that it is a non-selective cation channel.
  • the conductance of the channel is measured with various alkaline ions in the pipette solution, including Cs + , Na + , Rb + , K + , and Li + , always with equimolar K + in the bath solution.
  • Current-voltage data are fit to the GHK equation. Na + is shown to have a nearly equal slope conductance (32.6 pS) compared to K + (35.2 pS), but the slope conductance is reduced with other cations ( FIG. 3B ).
  • the permeability of the NC Ca-ATP channel of the present invention to anions, such as Cl ⁇ , is also assessed.
  • the bath solution is changed to equimolar K + gluconate.
  • the solution change resulted in a change in Erev ⁇ 0.5 mV, indicating that the NC Ca-ATP channel of the present invention is essentially impermeable to anions.
  • the permeability of the instant channel to divalent cations, Ca 2+ and Mg 2+ is also investigated ( FIG. 3C ).
  • potassium ion in the pipette solution is replaced with 75 mM Ca 2+ or Mg 2+ , inward currents are not detected.
  • Fit to the GHK equation gives best fit values for E rev , ⁇ 65 mV for Ca 2+ and Mg 2+ respectively, giving relative permeabilities with respect to K + of ⁇ 0.001, indicating that this channel is essentially impermeable to divalent cations.
  • NC Ca-ATP channel of the present invention discriminates very poorly among monovalent inorganic cations ( FIGS. 3A and B)
  • experiments are performed to determine the equivalent pore size of the channel by measuring channel permeability, relative to Cs + , for a wide range of organic cations.
  • single-channel current-voltage relations are plotted to obtain E rev for a number of organic cations. Permeability ratios are then derived from fits to the GHK equation.
  • the mean value of relative permeability measured is plotted against its hydrated molecular radius ( FIG. 3D , empty circles).
  • the permeability ratios define a smoothly declining series of values that are well fit by the Renkin equation.
  • the Renkin equation describes the permeation of a rigid sphere through a cylindrical pore. Renkin, 1955. Least-squares, fit to the equation, indicates an equivalent pore radius of 0.67 nm for the NC Ca-ATP channel of the present invention.
  • a 0.67 nm pore radius is similar to pore sizes of 6 A, found for the Ca 2+ channel (McCleskey and Almers, 1985) and 7.4 A, found for the nAChR channel (Adams et al., 1980). Junction potentials determined according to the methods described herein generally did not exceed 5 mV.
  • NC Ca-ATP channel is inhibited by intracellular ATP, based on the finding that this channel is turned on after depleting intracellular ATP by exposure to NaN 3 (See FIGS. 1B , 1 C, 1 D and 1 E) or to NaCN plus 2-deoxyglucose. This fact is supported by the observation that the NC Ca-ATP channel of the present invention is seldom observed in cell attached patches from healthy cells, but becomes evident in >90% of patches after conversion to an inside-out configuration.
  • the NC Ca-ATP channel is blocked by [ATP], in a dose-dependent manner.
  • ADP and AMP have no effect on the NC Ca-ATP channel activity in inside-out patches.
  • This in vitro assay for determining the concentration of the test compound which achieves a half-maximal inhibition of channel activity may be used to formulate dose in animal models to achieve a circulating plasma concentration range that includes the IC 50 .
  • the Ca 2+ concentration on the cytoplasmic side of the membrane is also found to regulate activity of the NC Ca-ATP channel of the present invention.
  • free [Ca 2+ ]i is ⁇ 30 nM, no channel activity is apparent.
  • [Ca 2+ ]i>30 nM the open probability (no ⁇ o) increases in accordance with the [Ca 2+ ]i, up to ⁇ 1 ⁇ M of [Ca 2+ ]i at which activity is near maximum.
  • FIG. 7A is a photograph of the gel showing the RT-PCR for SUR1 and SUR2.
  • FIG. 7B is a photograph of a gel showing the RT-PCR for Kir6.1 and Kir6.2.
  • Lanes 3 and 4 in FIGS. 7A and 7B show the RT-PCR for insulinoma cells.
  • Lanes 5 and 6 show the RT-PCR for reactive astrocytes.
  • Lane 1 in FIGS. 7A and 7B represents ladder size markers; Lane 2 in FIGS. 7A and 7B is a blank control.
  • FIG. 7A is a photograph of the gel showing the RT-PCR for SUR1 and SUR2.
  • FIG. 7B is a photograph of a gel showing the RT-PCR for Kir6.1 and Kir6.2.
  • Lanes 3 and 4 in FIGS. 7A and 7B show the RT-PCR for insulinoma cells.
  • Lanes 5 and 6 show the RT-PCR for reactive astrocytes.
  • Lane 1 in FIGS. 7A and 7B represents ladder
  • lanes 3 and 4 show the SUR1 and SUR2 experiments, respectively, in insulinoma cells. Insulinoma cells are known to express SUR1, but not SUR2. Lanes 5 and 6 in FIG. 7A show the SUR1 and SUR2 experiments in reactive astrocytes, respectively.
  • FIG. 7A shows that SUR1 mRNA is present in reactive astrocytes, as well as in the control insulinoma cells. SUR2 is absent in both cell types.
  • FIG. 7B lanes 3 and 4 show the Kir6.1 and Kir6.2 experiments in insulinoma cells, respectively. Kir6.1 is present in insulinoma cells, but Kir6.2 is not. Kir6 is the potassium channel associated with SUR1 in insulinoma cells. Lane 5 and 6 in FIG. 7B show that neither Kir6.1 nor Kir6.2 is present in reactive astrocytes. Therefore, reactive astrocytes express SUR1 mRNA, but Kir6.1 and Kir6.2 mRNA is absent from the cells.
  • SUR1 in reactive astrocytes combined with the regulation of the NC Ca-ATP channel in astrocytes by SUR antagonists indicates that SUR regulates the NC Ca-ATP channel of the present invention.
  • a characteristic feature of SUR-regulated K A ⁇ function is that tryptic digestion of the cytoplasmic face of the channel, but not its extracellular face causes loss of inhibition by sulfonylureas, without altering sensitivity to ATP and without changing the biophysical properties of the channel.
  • the effect of trypsin on NC Ca-ATP function is shown in FIG. 8 .
  • channel activity in the inside-out patch configuration is strongly inhibited by 1 pM glibenclamide.
  • Sulfonylurea compounds are known to modulate the sulfonylurea receptor.
  • a sulfonylurea receptor is generally associated with K ATP channels as a regulatory component, and is found in various tissues, including rat NRAs. Notably, the K ATP channels Kir6.1 and Kir6.2 are not present in rat NRAs ( FIG. 7B ). It is possible to activate the NC Ca-ATP channel with SUR ligand diazoxide in outside-out patches ( FIGS. 9A and 9B ). NaN 3 does not elicit channel activity in isolated membrane patches, indicating that it works via ATP depletion rather than any direct effect on the channel.
  • SUR1 blocking compounds such as glibenclamide and tolbutamide
  • SUR1 blocking compounds are known to have an inhibitory effect on K ATP channels.
  • the present invention arrives at the objects of the invention by providing a method in which the direct inhibitory effect of glibenclamide and tolbutamide on NC Ca-ATP channels is determined ( FIGS. 10 and 11 ). Inside-out patches are used to show the inhibitory effect of sulfonylureas. To ensure that no K+ channel, particularly K ATP is contributing to patch current, Cs+ is used as the charge carrier. Channel activity is profoundly diminished by the addition of 10 ⁇ M glibenclamide ( FIGS.
  • FIG. 10A the activity is shown to be due to a 35 pS cation channel, which is consistent with the NC Ca-ATP channel of the present invention ( FIG. 10C ).
  • Another sulfonylurea, tolbutamide is also shown to inhibit NC Ca-ATP channel activity ( FIGS. 11A and 11B ).
  • the NC Ca-ATP channel is blocked by the sulfonylureas in a dose-dependent manner.
  • half maximum inhibition EC 50
  • EC 50 half maximum inhibition
  • NC Ca-ATP channel of the present invention to blocking in NRAs with both of these sulfonylurea compounds corresponds closely to that reported in pancreatic p cells and in expression systems with SUR1, but not SUE.
  • This in vitro assay for determining the concentration of the test compound which achieves a half-maximal inhibition of channel activity may be used to formulate dose in animal models to achieve. a circulating plasma concentration range.
  • the NC Ca-ATP channel of the present invention exhibits two open states, with a shorter and a longer dwell time, each less than 10 ms.
  • FIG. 12 shows data from a patch exhibiting an open channel probability (n ⁇ Po) of 0.63, with open dwell time values ⁇ 0-1 ⁇ 0-2 and of 1.9 and 8.2 ms.
  • n ⁇ Po decreased to 0.44 and 0.09, respectively, but the open dwell time values are not appreciably affected by the drug.
  • Closed channel dwell times are increased in duration and frequency by tolbutamide ( FIGS. 12H and 12I ).
  • the channel of the present inventions exhibits a form of channel inhibition in which the blocking compound had no effect on open channel dwell times and a progressive increase in long closures.
  • This form of channel inhibition is similar to that produced by sulfonylureas acting on the K ATP channel in pancreatic ⁇ cells. See, Gillis et. al., 1989; Babeenko et. al., 1999).
  • Diazoxide is an SUR1 agonist or SUR1 activator.
  • SUR1 SUR1 activator
  • blebbing occurs even without ATP depletion
  • Diazoxide therefore, opens the channel directly without ATP depletion by activating SUR1.
  • addition of NaN 3 does not cause blebbing, even after 30 minutes.
  • activation of NC Ca-ATP channel by ATP depletion or by the channel opener, diazoxide can result in blebbing and swelling of NRAs, and that swelling can be prevented by blocking the channel with glibenclamide.
  • glibenclamide protects from the opening of the NC Ca-ATP channel following ATP depletion, and that opening of this channel is responsible for cell blebbing.
  • the antagonist used in the methods of the present invention includes a compound that interferes with NC Ca-ATP function.
  • the effect of an antagonist is observed as a blocking of NC Ca-ATP current in conditions under which the channel has been activated and current can be measured in the absence of the antagonist.
  • agents that block SUR1 also include compounds that are structurally unrelated to sulfonylureas.
  • SUR1 blockers include a class of insulin secretagogues compounds that bind to the SUR, which were identified and developed for the treatment of type 2 diabetes.
  • the benzamido derivatives: repaglinide, nateglinide, and meglitinide represent one such class of insulin secretagogues, that bind to the SUR.
  • Nateglinide is an amino acid derivative.
  • imidazoline derivatives have been identified that interact with the sulfonylurea receptor (SUR) 1 subunit such as midaglizole (KAD-1229), LY397364 and LY389382.
  • compounds that preferentially block SUR1, but not SUR2 are used in the method of the present invention.
  • Such compounds include tolbutamide and gliclazide.
  • the following compounds block both SUR1 and SUR2: glibenclamide, glimepiride, repaglinide, and meglitinide.
  • administration is combined with MgADP, which has been show to produce an apparent increase of sulfonylurea efficacy on channels containing SUR1, but not SUR2.
  • NC Ca-ATP activation by ATP depletion initiates necrosis of reactive astrocytes that express this channel
  • studies are conducted to determine if glibenclamide is capable of protecting reactive astrocytes from cell death by inhibiting NC Ca-ATP channel activity via its action on SUR1. Two types of cell death, apoptosis and necrosis, are assessed following ATP depletion.
  • NC Ca-ATP channel activation of NC Ca-ATP channel is responsible for necrotic death of NRAs following ATP depletion, and that glibenclamide can prevent this form of cell death.
  • NRAs preparation of freshly isolated NRAs was further purified by removal of RBCs, as described herein to provide a cell population having ⁇ 1% RBCs. Over 95% of cells had resting potentials near EK, suggesting that the enzymatic dissociation method had not appreciably harmed the cells. Over 95% of cells are positive for the astrocyte marker, glial fibrillary acidic protein (GFAP) as determined by immunofluorescence.
  • GFAP glial fibrillary acidic protein
  • the NRAs When examined by phase microscopy, the NRAs are of various sizes, ranging from 11-45 pms in diameter, some of which are phase bright and others are phase dark. A subgroup of phase bright cells had multiple short but distinct cell processes that are shorter than the cell soma. In this Example, only larger ( ⁇ 30 ⁇ m diameter), phase bright cells with short processes ( ⁇ 1 cell length) are studied. This population of NRAs reliably express the NC Ca-ATP channels.
  • the cells are examined by propidium iodide (PI) staining for evidence of cellular membrane permeabilization, an indication of early oncotic or necrotic cell death. See, Barros et al., 2001.
  • the cells are also examined by fluorescein-tagged annexin V binding for evidence of externalization of the phosphoaminolipid phosphotidylserine from the inner face of the plasma membrane to the outer surface, an early indication of apoptosis. See, Clodi et al., 2000; Rucker-Martin et al., 1999. Staining procedure are conducting according to manufacture directions (Vybrant Apoptosis Assay Kit 2, Molecular Probes).
  • FIG. 15A The fluorescence microscopy photos shown in FIG. 15A show that under baseline (control) conditions, both annexin V-positive and PI-positive cells (photos a and d, respectively) are rare in the cell isolates.
  • the number of PI-positive cells increased substantially (p ⁇ 0.05) ( FIG. 15A at photo b and FIG. 15B ). This indicates that ATP depletion triggers necrotic death in these cells.
  • Na azide treatment caused the number of annexin V-positive cells to increase slightly; the increase not being statically significant (p>0.05) ( FIG. 15A at photo e and FIG. 15C ). This indicates that apoptotic death was not a major endpoint of ATP depletion in these cells.
  • Pretreatment of cells with glibenclamide (1 ⁇ M) at the time of administration of Na aide dramatically decreased the number of PI-positive cells (p ⁇ 0.05; FIG. 15A at photo c and FIG. 15B ), indicating significant protection from necrotic death following ATP depletion.
  • the number of NRAs undergoing apoptotic death also decreased with glibenclamide, as indicated by annexin V labeling ( FIG. 15A at photo f and FIG. 15C ), but values for this group were not significantly different.
  • NC Ca-ATP channel is involved in the mechanism of the necrotic cell death of reactive astrocytes.
  • This Example shows that necrotic, rather than apoptotic, cell death is the principal endpoint of ATP depletion in these cells. Therefore, ATP depletion by Na azide initiates cell death by removal of the ATP block of the NC Ca-ATP channel, thus initiating oncotic cell swelling. Involvement of this channel in oncotic cell swelling is confirmed by showing that necrotic death can also be induced by diazoxide, the channel opener that activates the NC Ca-ATP channel in these cells, and could be blocked by glybenclamide, which prevents opening of the NC Ca-ATP channel.
  • the involvement of the NC Ca-ATP channel in cell death of reactive astrocytes provides a mechanism and target of death in these cells, as well as the importance of blocking the NC Ca-ATP channel to prevent the death of reactive astrocytes, which occurs in traumatic brain injury.
  • NC Ca-ATP channels blocking compounds can be identified by a method in which the direct inhibitory effect of the test compound on NC Ca-ATP channels is determined. Inside-out patches are used to show the inhibitory effect of the compound. To ensure that no K + channel, particularly K ATP is contributing to patch current, Cs+ is used as the charge carrier. Compounds that profoundly diminish channel activity, and the activity is shown to be due to a 35 pS cation channel, such a compound is identified as a compound that blocks the NC Ca-ATP channels and is capable of inhibiting neuronal cell swelling and brain swelling. Varying concentrations of the compound are used to determine whether the NC Ca-ATP channel is blocked by the compound in a dose-dependent manner.
  • the concentration at which half maximum inhibition (EC 50 ) is observed and the concentration at which channel activity is completely lost are determined.
  • the sensitivity of the NC Ca-ATP channel of the present invention to blocking in NRAs with the test compound can be compared.
  • This in vitro assay for determining the concentration of the test compound which achieves a half-maximal inhibition of channel activity may be used to formulate dose in animal models to achieve a circulating plasma concentration range.
  • the concentration of the test compound which achieves a half-maximal inhibition of channel activity is used to formulate dose in animal models to achieve a circulating plasma concentration range.
  • the dose of test compound that achieves a circulating plasma concentration range calculated by methods known in the art is administered to an animal having brain injury or cerebral ischemia.
  • the epidural pressure and/or intracranial pressure of the animal is measured, such as by using a microballoon, to quantitatively monitor brain swelling.
  • the swelling can be monitored by magnetic resonance (MR) imaging.
  • MR magnetic resonance
  • a compound that provided diminishes brain swelling, as compared to controls, is identified as a compound capable of inhibiting neuronal cell swelling and brain swelling. Varying concentrations of the compound are used to determine whether the compound delivers efficacy in a dose-dependent manner. The dose at which half maximum inhibition is observed and the concentration at which brain swelling is most quickly alleviated are determined. Formulations are produced comprising the optimal effective dose of the test compound for preventing, inhibiting, or diminishing brain swelling, along with a pharmaceutically acceptable carrier.
  • the present invention provides a previously unknown ion channel found in mammalian neural cells that plays a role in cell swelling.
  • the present invention further provides a method of screening for antagonists to the channel and a new use for antagonists to the channel, including sulfonylurea compounds such as glibenclamide and tolbutamide, as a treatment for brain swelling in mammals.
  • Such compounds may act as antagonists or agonists of NCCa-Ap channel activity.
  • antagonists that block and/or inhibit the permeability of the NC Ca-ATP channel are utilized in methods for treating neural cell swelling and/or brain swelling.
  • the cell based assays use neural cells that express the NC Ca-ATP channel, preferably a functional NC Ca-ATP channel; the preferred cells are “MS.
  • the non-cell based assay systems include membrane preparations that express the NC Ca-ATP channel, preferably a functional NC Ca-ATP channel.
  • Cell-based assays include, but are not limited to, compound binding assays, microscopic observation of cell status (normal, blebbing, swelling), and measuring channel currents both before and after exposure to compound.
  • Compositions comprising membrane preparations expressing the NC Ca-ATP channel may be used to identify compounds that interact with, bind to, block or open the NC Ca-ATP channel or SUR1.
  • NC Ca-ATP channel or “expresses the NC Ca-ATP channel” means having a functional NC Ca-ATP channel.
  • functional NC Ca-ATP channel as used herein means an NC Ca-ATP channel capable of being detected.
  • One preferred method of detecting the NC Ca-ATP channel is by determining, in vitro or in vivo, whether the channel is open, closed and/or blocked.
  • NRAs that express the NC Ca-ATP channel are used to produce the membrane preparation.
  • Methods for producing membranes from whole cells and tissues are well known in the art.
  • One such method produces purified cell membranes in the form of a purified microsomal fraction isolated from disrupted cells or a tissue sample by discontinuous sucrose gradient centrifugation.
  • membranes comprised of cell-attached patches, inside-out patches, or outside-out patches.
  • tissue sample expressing NC Ca-ATP channels is brain tissue adjacent to brain injury.
  • the membranes preparations are used in a number of assays, including, but not limited to measuring channel currents, both before and after exposure to compound; and in vitro binding assays.
  • assays including, but not limited to measuring channel currents, both before and after exposure to compound; and in vitro binding assays.
  • the experimental conditions for such assays to determine and quantify the status of the NC Ca-ATP channel are described throughout the instant specification, including binding assay conditions, bath compositions, pipette solutions, concentrations of ATP and Ca 2+ required, membrane voltage, membrane potentials, compound quantity ranges, controls, etc.
  • Binding assays and competitive binding assays employ a labeled ligand or antagonist of the NC Ca-ATP channel.
  • labeled Glibenclamide such as FITC-conjugated glibenclamide or radioactively labeled glibenclamide is bound to the membranes and assayed for specific activity; specific binding is determined by comparison with binding assays performed in the presence of excess unlabelled antagonist.
  • membranes are incubated with a labeled compound shown to block this channel, in either the presence or absence of test compound.
  • Compounds that block the NC Ca-ATP channel and compete with the labeled compound for binding to the membranes will have a reduced the signal, as compared to the vehicle control samples.
  • the screens may be designed to identify compounds that compete with the interaction between NC Ca-ATP channel and a known (previously identified herein) NC Ca-ATP channel antagonist or SUR1 antagonist, such as glibenclamide.
  • the known NC Ca-ATP channel antagonist or SUR1 antagonist is labeled and the test compounds are then assayed for their ability to compete with or antagonize the binding of the labeled antagonist.
  • the assays described herein can be used to identify compounds that modulate or affect NC Ca-ATP channel activity.
  • compounds that affect NC Ca-ATP channel activity include but are not limited to compounds that bind to the NC Ca-ATP channel or SUR1, inhibit binding of identified blockers or ligands (such as glibenclamide), and either open/activate the channel (agonists) or block/inhibit the channel (antagonists).
  • Assays described can also identify compounds that modulate neural cell swelling (e.g., compounds which affect other events involved in neural cell swelling that are activated by ligand binding to or blocking of the NC Ca-ATP channel).
  • the compounds for screening in accordance with the invention include, but are not limited to organic compounds, peptides, antibodies and fragments thereof, peptidomimetics, that bind to the NC Ca-ATP channel and either open the channel (i.e., agonists) or block the channel (i.e., antagonists).
  • compounds that block the channel are preferred.
  • Agonists that open or maintain the channel in the open state include peptides, antibodies or fragments thereof, arid other organic compounds that include the SUR1 subunit of the NC Ca-ATP channel (or a portion thereof) and bind to and “neutralize” circulating ligand for SUR1.
  • libraries of known compounds can be screened, including natural products or synthetic chemicals, and biologically active materials, including proteins, for compounds which are inhibitors or activators.
  • a compound is an NC Ca-ATP antagonist, which includes an NC Ca-ATP channel inhibitor, an NC Ca-ATP channel blocker, a SUR1 antagonist, SUR1 inhibitor, and/or a compound capable of reducing the magnitude of membrane current through the channel.
  • Compounds may include, but are not limited to, small organic or inorganic molecules, compounds available in compound libraries, peptides such as, for example, soluble peptides, including but not limited to members of random peptide libraries; (see, e.g., Lam, K. S. et al., 1991, Nature 354: 82-84; Houghten, R. et al., 1991, Nature 354: 84-86), and combinatorial chemistry-derived molecular library made of D- and/or L-configuration amino acids, phosphopeptides (including, but not limited to, members of random or partially degenerate, directed phosphopeptide libraries; see, e.g., Songyang, Z.
  • peptides such as, for example, soluble peptides, including but not limited to members of random peptide libraries; (see, e.g., Lam, K. S. et al., 1991, Nature 354: 82-84; Houghten, R. et
  • antibodies including, but not limited to, polyclonal, monoclonal, humanized, anti-idiotypic, chimeric or single chain antibodies, and FAb, F(ab′).sub.2 and FAb expression library fragments, and epitope-binding fragments thereof).
  • Other compounds which can be screened in accordance with the invention include but are not limited to small organic molecules that are able to cross the blood-brain barrier, gain entry into an appropriate neural cell and affect the expression of the NC Ca-ATP channel gene or some other gene involved in the NC Ca-ATP channel activity (e.g., by interacting with the regulatory region or transcription factors involved in gene expression); or such compounds that affect the activity of the NC Ca-ATP channel or the activity of some other intracellular factor involved in the NC Ca-ATP channel activity.
  • Computer modeling and searching technologies permit identification of compounds, or the improvement of already identified compounds, that can modulate NC Ca-ATP channel activity or expression. Having identified such a compound or composition, the active sites or regions are identified. Such active sites might typically be ligand binding sites.
  • the active site can be identified using methods known in the art including, for example, from study of complexes of the relevant compound or composition with other ligands, from the amino acid sequences of peptides, or from the nucleotide sequences of nucleic acids. Chemical or X-ray crystallographic methods can be used to study complexes of the relevant compound to find the active site. The three dimensional geometric structure of the active site is determined.
  • the methods of computer based numerical modeling can be used to complete the structure or improve its accuracy.
  • Any recognized modeling method may be used, including parameterized models specific to particular biopolymers such as proteins or nucleic acids, molecular dynamics models based on computing molecular motions, statistical mechanics models based on thermal ensembles, or combined models.
  • standard molecular force fields representing the forces between constituent atoms and groups, are necessary, and can be selected from force fields known in physical chemistry.
  • the incomplete or less accurate experimental structures can serve as constraints on the complete and more accurate structures computed by these modeling methods.
  • candidate modulating compounds can be identified by searching databases containing compounds along with information on their molecular structure. Such a search seeks compounds having structures that match the determined active site structure and that interact with the groups defining the active site. Such a search can be manual, but is preferably computer assisted. These compounds found from this search are potential NC Ca-ATP channel modulating, preferably blocking, compounds.
  • these methods can be used to identify improved modulating compounds from an already known modulating compound or ligand.
  • the composition of the known compound can be modified and the structural effects of modification can be determined using the experimental and computer modeling methods described above applied to the new composition.
  • the altered structure is then compared to the active site structure of the compound to determine if an improved fit or interaction results. In this manner systematic variations in composition, such as by varying side groups, can be quickly evaluated to obtain modified modulating compounds or ligands of improved specificity or activity.
  • CHARMm performs the energy minimization and molecular dynamics functions.
  • QUANTA performs the construction, graphic modeling and analysis of molecular structure.
  • QUANTA allows interactive construction, modification, visualization, and analysis of the behavior of molecules with each other.
  • a number of articles review computer modeling of drugs interactive with specific proteins, such as Rotivinen, et al.) 1988, Acta Pharmaceutical Fennica 97: 159-166); Ripka (1988 New Scientist 54-57); McKinaly and Rossmann (1 989, Annu. Rev. Pharmacol. Toxicol.
  • Compounds identified via assays such as those described herein may be useful, for example, in elaborating the biological function of the NC Ca-ATP channel and for relief of brain swelling.
  • Assays for testing the efficacy of compounds identified in the cellular screen can be tested in animal model systems for brain swelling.
  • animal models may be used as test substrates for the identification of drugs, pharmaceuticals, therapies and interventions which may be effective in treating brain swelling.
  • animal models of brain swelling such as brain injury
  • the response of the animals to the exposure may be monitored using visual means (e.g., radiological, CAT, MRI), measurement of intracranial pressure, and/or the reversal of symptoms associated with brain swelling.
  • visual means e.g., radiological, CAT, MRI
  • measurement of intracranial pressure e.g., intracranial pressure
  • any treatments which reverse any aspect of brain swelling-associated symptoms should be considered as candidates for brain swelling therapeutic intervention.
  • Dosages of test agents may be determined by deriving dose-response curves, as discussed herein.
  • the present invention is useful in the treatment or alleviation of neural cell swelling and death and brain swelling, especially those brain insults related to traumatic brain injury, central or peripheral nervous system damage, cerebral ischemia, such as stroke, or complications involving and/or stemming from edema, injury, or trauma.
  • damage or complications may be characterized by an apparent brain damage or aberration, the symptoms of which can be reduced by the methods of the present invention including the administration of an effective amount of the active compounds or substances described herein.
  • the administration of effective amounts of the active compound can block the channel, which if remained open leads to neural cell swelling and cell death.
  • a variety of antagonists to SUR1 are suitable for blocking the channel.
  • SUR1 antagonists include, but are not limited to glibenclamide, tolbutamide, repaglinide, nateglinide, meglitinide, midaglizole, LY397364, LY3 89382, gliclazide, glimepiride, MgADP, and combinations thereof.
  • the SUR1 antagonists is selected from the group consisting of glibenclamide and tolbutamide. Still other therapeutic “strategies” for preventing neural cell swelling and cell death can be adopted including, but not limited to methods that maintain the neural cell in a polarized state and methods that prevent strong depolarization.
  • reactive astrocytes When reactive astrocytes are strongly depolarized due to opening of the NC Ca-ATP channel, they undergo blebbing and swelling and eventually sustain necrotic cell death. As stated above, when reactive astrocytes are strongly depolarized due to opening of a non-selective channel that is sensitive to Ca 2+ and ATP (NC Ca-ATP channel), they undergo blebbing and swelling and eventually sustain necrotic cell death. The death of these reactive astrocytes can be prevented if strong depolarization can be prevented, in other words, if the cells can be maintained in a polarized state.
  • One potential way of maintaining the NRAs in a polarized state is to open the Kir2.3 channel.
  • NRAs are exposed to the Kir2.3 channel opener, Tenidap, to maintain Kir2.3 channels open.
  • Native reactive astrocytes freshly harvested from adult rat brains after injury are exposed to Tenidap to evaluate the drug's ability to open the Kir2.3 channel in these cells.
  • type 1 reactive (R1) astrocytes are harvested and used in this assay.
  • One of the subtypes of reactive astrocytes is the type R1 astmcyte.
  • Type R1 astrocytes comprise the largest population of recoverable astrocytes at the site of brain injury. They are characteristically located in the region of tissue surrounding the injury site, many of which are found to have migrated into the injury site itself. See, Perillan, et al., 1999.
  • the reactive astrocytes that are part of the cellular response to TBI and stroke are comprised of at least two subtypes.
  • One of the subtypes of reactive astrocytes is the type R1 astrocyte.
  • Type R1 astrocytes comprise the largest population of recoverable astrocytes at the site of brain injury. They are characteristically located in the region of tissue surrounding the injury site, with many of these cells also being found to have migrated into the injury site itself. See, Perillan, et al. 1999.
  • Type R1 astrocytes are the predominant type of reactive astrocyte in the NRA preparations.
  • Type R1 astrocytes express two critically important ion channels in their cell membrane: (a) the Kir2.3 channel, which is present in cultured as well as freshly isolated cells; and (b) the NC Ca-ATP channel, which is present only in freshly isolated reactive astrocytes and lost shortly after culturing.
  • the Kir2.3 is an inward rectifier channel that is critically important for maintaining the cell polarized to a normal resting potential near the potassium reversal potential ( ⁇ 75 mV). When this channel is inactivated or inhibited, the cell depolarizes to a potential near the chloride reversal potential ( ⁇ 25 mV).
  • NC Ca-ATP channel Characteristic features of the NC Ca-ATP channel are: 1) it is a non-selective cation channels that allows passage of Nay K and other monovalent cations quite readily; 2) it is activated by an increase in intracellular calcium, and/or by a decrease in intracellular ATP; and 3) it is regulated by sulfonylurea receptor type 1 (SUR1). SUR1 had been considered to be associated exclusively with K ATP channels, such as those found in pancreatic cells.
  • This influx of Na increases the osmotic load within the cell, and as a result, H 2 0 enters the cell to equilibrate the osmotic load.
  • the result is an excess of Na and H 2 0 intracellularly, a pathological response that produces cell blebbing and cell swelling and that is known as cytotoxic edema. Left unchecked, this pathological response eventually leads to cell death. As disclosed herein, this cell death is mostly necrotic cell death but to a lesser extent, apoptotic cell death as well.
  • a number of approaches may be used to meliorate brain swelling due to cytotoxic edema.
  • One currently used treatment for treating patients in relevant clinical situations is based on increasing extracellular osmolarity to reduce the driving force for influx of H 2 0. This strategy also reduces blebbing in isolated cells.
  • a more specific strategy to reduce cytotoxic edema is inactivating or blocking the NC Ca-ATP channel that is primarily responsible for the influx of Na that draws H 2 0 into the cell and that actually causes cytotoxic edema.
  • One highly selective approach to inactivating this channel is to exploit the unique relationship between the channel and the controlling regulatory subunit, SUR1.
  • SUR1 the controlling regulatory subunit
  • a variety of drugs have been developed that interact with SUR1 in pancreatic p cells to block the K ATP channel in those cells and thereby treat diabetes. Some of these drugs belong to the class of agents called sulfonylureas.
  • drugs that block the K ATP channel are highly effective at blocking the NC Ca-ATP channel in type R1 astrocytes.
  • Drugs capable NC Ca-ATP channel blocking in NRAs (a) prevents cell blebbing in response to ATP depletion, (b) significantly reduces cell death following ATP depletion.
  • the use of glybenclamide to treat brain swelling in an animal suffering from stroke or brain injury is described herein.
  • Yet another strategy to oppose the effect of the NC Ca-ATP channel and reduce cytotoxic edema would be to counteract depolarization of the cell that accompanies opening of the NC Ca-ATP channel.
  • One way to accomplish this is to enhance opening of the Kir2.3 channels that are also present in these cells.
  • An anti-inflammatory compound, Tenidap is an opener of Kir2.3 channels. See, Popp et al., 1992; Liu et al., 2002. [OO1SO] Tenidap is evaluated for its ability to reduce cell blebbing and swelling and necrotic cell death in response to ATP depletion in the isolated cells as well as in situ in injured rat brain.
  • Tenidap opens the Kir2.3 channels in type R1 astrocytes, using methods similar to those described herein for evaluating the status of the NC Ca-ATP channel. Results from such experiments that show Tenidap to open Kir2.3 channels in type R1 astrocytes, and reduce cell blebbing and cell death in response to ATP depletion would indicate the usefulness of Tenidap in treating brain swelling and cytotoxic edema resulting from TBI or cerebral ischemia.
  • the effective amount of Tenidap is that amount capable of reducing brain swelling or cerebral ischemia due to the drug's ability to inhibit neural cell swelling and necrotic cell death.
  • SUR1 blockers are likely to be the most specific, reliable provide the fewest untoward side effects. Further, a combination of treatments including use of osmotic diuretics, NC Ca-ATP channel blockers such glybenclamide and Kir2.3 channel openers such as Tenidap may provide better efficacy in ameliorating cytotoxic edema and reducing morbidity and mortality in brain injury and stroke.
  • a preferred compound is Tenidap.
  • the formulation may provide a daily dose of Tenidap that is from about 10 mg/day to about 500 mg/day, or, when administered directly to the brain the daily dose of Tenidap is from about 500 mg/day to 1.5 gms/day or greater.
  • compositions comprising the active substances disclosed herein.
  • these compositions include pharmaceutical compositions comprising a therapeutically effective amount of one or more of the active compounds or substances along with a pharmaceutically acceptable carrier.
  • the term “pharmaceutically acceptable” carrier means a non-toxic, inert solid, semi-solid liquid filler, diluent, encapsulating material, formulation auxiliary of any type, or simply a sterile aqueous medium, such as saline.
  • sugars such as lactose, glucose and sucrose, starches such as corn starch and potato starch, cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt, gelatin, talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol, polyols such as glycerin, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate, agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline, Ringer's solution; ethyl
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
  • antioxidants examples include, but are not limited to, water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfite, sodium metabisulfite, sodium sulfite, and the like; oil soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, aloha-tocopherol and the like; and the metal chelating agents such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfite, sodium metabisulfite, sodium sulfite, and the like
  • oil soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (B
  • a “therapeutically effective amount” or simply “effective amount” of an active compound is meant a sufficient amount of the compound to treat or alleviate the brain swelling at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the active compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the brain injury or ischemia; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coinciding with the specific compound employed; and like factors well known in the medical arts.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell assays or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell based assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC50 i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • the total daily dose of the active compounds of the present invention administered to a subject in single or in divided doses can be in amounts, for example, from 0.01 to 25 mg/kg body weight or more usually from 0.1 to 15 mg/kg body weight.
  • Single dose compositions may contain such amounts or submultiples thereof to make up the daily dose.
  • treatment regimens according to the present invention comprise administration to a human or other mammal in need of such treatment from about 1 mg to about 1000 mg of the active substance(s) of this invention per day in multiple doses or in a single dose of from 1 mg, 5 mg, 10 mg, 100 mg, 500 mg or 1000 mg.
  • the typical anti-diabetic dose of oral or IV glibenclamide is about 2.5 mg/kg to about 15 mg/kg per day; the typical anti-diabetic dose of oral or IV tolbutamide is about to 0.5 gm/kg to about 2.0 gm/kg per day; the typical anti-diabetic dose for oral gliclazide is about 30 mg/kg to about 120 mg/kg per day; however, much larger doses may be required to block neural cell swelling and brain swelling.
  • a formulation containing an effective amount of a compound that blocks the NC Ca-ATP channel and a pharmaceutically acceptable carrier may contain from about 0.1 to about 100 grams of tolbutamide or from about 0.5 to about 150 milligrams of glibenclamide.
  • a method of alleviating the negative effects of traumatic brain injury or cerebral ischemia stemming from neural cell swelling in a subject by administering to the subject a formulation containing an effective amount of a compound that blocks the NC Ca-ATP channel and a pharmaceutically acceptable carrier.
  • traumatic brain injury or cerebral ischemia such as stroke
  • cerebral hypoxia it may be important to maintain a fairly high dose of the active agent to ensure delivery to the brain of the patient, particularly early in the treatment.
  • traumatic brain injury or cerebral ischemia such as stroke
  • the method of the present invention is employed to treat conditions involving bleeding in the brain, such as traumatic brain injury or cerebral ischemia (such as stroke), delivery via the vascular system is available and the compound is not necessarily required to readily cross the blood-brain barrier.
  • the compounds of the present invention may be administered alone or in combination or in concurrent therapy with other agents which affect the central or peripheral nervous system, particularly selected areas of the brain.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs containing inert diluents commonly used in the art, such as water, isotonic solutions, or saline.
  • Such compositions may also comprise adjuvants, such as wetting agents; emulsifying and suspending agents; sweetening, flavoring and perfuming agents.
  • sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulation can be sterilized, for example, by filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions, which can be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.
  • the most common way to accomplish this is to inject a suspension of crystalline or amorphous material with poor water solubility.
  • the rate of absorption of the drug becomes dependent on the rate of dissolution of the drug, which is, in turn, dependent on the physical state of the drug, for example, the crystal size and the crystalline form.
  • Another approach to delaying absorption of a drug is to administer the drug as a solution or suspension in oil.
  • Injectable depot forms can also be made by forming microcapsule matrices of drugs and biodegradable polymers, such as polylactide-polyglycoside.
  • the rate of drug release can be controlled.
  • biodegradable polymers include polyorthoesters and polyanhydrides.
  • the depot injectables can also be made by entrapping the drug in liposomes or microemulsions, which are compatible with body tissues.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable non-irritating excipient, such as cocoa butter and polyethylene glycol which are solid at ordinary temperature but liquid at the rectal temperature and will, therefore, melt in the rectum and release the drug.
  • a suitable non-irritating excipient such as cocoa butter and polyethylene glycol which are solid at ordinary temperature but liquid at the rectal temperature and will, therefore, melt in the rectum and release the drug.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, gelcaps and granules.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such as magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings and other release-controlling coatings.
  • compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferably, in a certain part of the intestinal tract, optionally in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention further include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • Transdermal patches have the added advantage of providing controlled delivery of active compound to the body.
  • dosage forms can be made by dissolving or dispersing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • the method of the present invention employs the compounds identified herein for both in vitro and in vivo applications.
  • the invention compounds can be incorporated into a pharmaceutically acceptable formulation for administration. Those of skill in the art can readily determine suitable dosage levels when the invention compounds are so used.
  • suitable dosage levels refers to levels of compound sufficient to provide circulating concentrations high enough to effectively block the NC Ca-ATP channel and prevent or reduce neural cell swelling in vivo.
  • compositions comprising at least one SUR1 antagonist compound (as described above), and a pharmaceutically acceptable carrier are contemplated.
  • Exemplary pharmaceutically acceptable carriers include carriers suitable for oral, intravenous, subcutaneous, intramuscular, intracutaneous, and the like administration. Administration in the form of creams, lotions, tablets, dispersible powders, granules, syrups, elixirs, sterile aqueous or non-aqueous solutions, suspensions or emulsions, and the like, is contemplated.
  • suitable carriers include emulsions, solutions, suspensions, syrups, and the like, optionally containing additives such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents, and the like.
  • suitable carriers include sterile aqueous or non-aqueous solutions, suspensions, or emulsions.
  • suitable carriers include sterile aqueous or non-aqueous solutions, suspensions, or emulsions.
  • non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate.
  • Such dosage forms may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. They may be sterilized, for example, by filtration through a bacteria-retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They can also be manufactured in the form of sterile water, or some other sterile injectable medium immediately before use.
  • the active compound is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers
  • the specification may have presented the method and/or process of the present invention as a particular sequence of steps. However, to the extent that the method or process does not rely on the particular order of steps set forth herein, the method or process should not be limited to the particular sequence of steps described. As one of ordinary skill in the art would appreciate, other sequences of steps may be possible. Therefore, the particular order of the steps set forth in the specification should not be construed as limitations on the claims. In addition, the claims directed to the method and/or process of the present invention should not be limited to the performance of their steps in the order written, and one skilled in the art can readily appreciate that the sequences may be varied and still remain within the spirit and scope of the present invention.
  • the specification may have presented the method and/or process of the present invention as a particular sequence of steps. However, to the extent that the method or process does not rely on the particular order of steps set forth herein, the method or process should not be limited to the particular sequence of steps described. As one of ordinary skill in the art would appreciate, other sequences of steps may be possible. Therefore, the particular order of the steps set forth in the specification should not be construed as limitations on the claims. In addition, the claims directed to the method and/or process of the present invention should not be limited to the performance of their steps in the order written, and one skilled in the art can readily appreciate that the sequences may be varied and still remain within the spirit and scope of the present invention.

Abstract

A composition comprising a novel Ca2+-activated, [ATP]i-sensitive nonspecific cation (NCCa-ATP) channel is described. The channel is found in mammalian neural cells and exhibits a different sensitivity to block by various adenine nucleotides, and is activated by submicromolar [Ca]i. The NCCa-ATP channel is activated under conditions of ATP depletion, which causes severe cell depolarization, followed by cell swelling. The NCCa-ATP channel is regulated by a sulfonylurea receptor and is inhibited by sulfonylurea compounds glibenclamide and tolbutamide. Methods employing compositions comprising the NCCa-ATP channel to screen for compounds that block the channel and the use of such antagonists as therapeutics in preventing brain swelling and damage are described. In addition, methods employing compositions comprising the Kir2.3 channel to screen for compounds that open the channel and the use of such antagonists as therapeutics in preventing brain swelling and damage are described.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a Continuation of U.S. patent application Ser. No. 11/099,322, filed on Apr. 5, 2005, which is a Divisional of U.S. patent application Ser. No. 10/391,561, filed on Mar. 20, 2003, which claims priority to U.S. Provisional Patent Application No. 60/365,933, filed on Mar. 20, 2002, all of which applications are incorporated by reference herein in their entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to a novel ion channel found in neural cells which participates in the cation flux involved in cell swelling. The invention also provides a method of screening for compounds that inhibit the activity of the ion channel. Methods to screen for and identify antagonists of the NCCa-ATP channel are provided. The invention further provides therapeutic methods for using compounds and compositions that inhibit the ion channel activity to inhibit or prevent the swelling of neural cells in brain. It has been discovered that neural cell swelling is mediated by the opening of a novel non-selective monovalent cationic ATP sensitive channel (the NCCa-ATP channel) and that this channel is coupled to sulfonylurea receptor type 1. Moreover, it has been found that neural cell swelling and cell death, particularly astrocyte swelling, can be inhibited by blocking the NCCa-ATP channel of the present invention, particularly by antagonizing receptors coupled to this channel, such as antagonizing the SUR1. The invention also encompasses the use of such compounds and compositions, that modulate NCCa-ATP channel activity to treat brain swelling. The present invention relates to methods for the treatment of brain swelling that results from brain trauma or cerebral ischemia, due to neural cell swelling and cell death.
  • BACKGROUND OF THE INVENTION
  • Following traumatic brain injury and stroke, the normal response of the surrounding brain is to mount a cellular response that includes formation of reactive astrocytes that are believed to be important to “contain” and “clean-up” the injury site. Swelling of neural cells is part of the cytotoxic or cell swelling response that characterizes brain damage in cerebral ischemia and traumatic brain injury, and is a major cause of morbidity and mortality. See, Staub et al., 1993; Kimelberg et al., 1995. A number of mediators have been identified that initiate swelling of neural cells, including elevation of extracellular K+, acidosis, release of neurotransmitters and free fatty acids. See, Kempski et al., 1991; Rutledge and Kimelberg, 1996; Mongin et al., 1999. Cytotoxic edema is a well-recognized phenomenon clinically that causes brain swelling, which worsens outcome and increases morbidity and mortality in brain injury and stroke.
  • Mechanisms underlying apoptotic death of reactive astrocytes have been studied. See, Tanaka et al., 2000; Yu et al., 2001. The mechanisms responsible for necrotic cell death have not been characterized. Apoptotic cell death is preceded by cell shrinkage and net loss of K. See, Yu et al., 1997; Yu et al., 1999. By contrast, in necrotic cell death, the plasma membrane is ruptured, causing cytosolic contents to be released and thereby triggering tissue inflammation. See, Leist and Nicotera, 1997. Necrotic cell death may be more deleterious to nearby viable tissues, given the secondary inflammatory damage that is initiated.
  • Necrotic cell death is initiated by osmotic swelling following influx of Na+, the major extracellular osmolyte. In most cell types, accumulation of Na+ intracellularly is regarded as a passive process that does not require activation of specific effectors but that is due instead to defective outward Na′ pumping under conditions of low [ATP]i. See, Leist and Nicotera, 1997; Trump et al., 1997. Cell blebbing or swelling, an indication of intracellular Na′ overload, is generally regarded as an early sign of necrotic cell death. See, Leist and Nicotera, 1997; Majno and Joris, 1995.
  • Inhibition of ATP synthesis or ATP depletion also causes neural cell swelling, blebbing and, if sufficiently severe, plasma membrane disruption and cell death. See, Jurkowitz-Alexander et al., 1993. The mechanisms of neural cell swelling associated with ATP-depletion remained incompletely characterized. See, Lomneth and Gruenstein, 1989; Juurlink et al., 1992; Rose et al., 1998.
  • One potential mechanism would be changes in Na+ and K+ concentration due to inhibition of the Na+/K+-ATPase pump. However, an equivalent degree of osmotic swelling induced by ouabain-mediated inhibition of the Na+/K+-ATPase pump in neural cells does not produce large depolarization, blebbing or cell death. See, Jurkowitz-Alexander et al., 1992; Brismar and Collins, 1993. Failure of the Na+/K+-ATPase pump, therefore, is not the mechanism critical to swelling of neural cells. None of these studies have identified the cellular mechanism instrumental in the cell swelling that is associated with brain damage in cerebral ischemia and traumatic brain injury.
  • One subtype of ATP sensitive cation channel is the non-selective cation channel, which are channels that are sensitive to Ca2+ and ATP. More specifically, non-selective cation channels are activated by intracellular Ca2+ ([Ca2+]I and inhibited by intracellular ATP ([ATP]i). Although Ca2+ and ATP sensitive cation channels had been identified in a number of non-neural cell types, they have not been identified in astrocytes or any other neural cells. See, Sturgess et al., 1987; Gray and Argent, 1990; Rae et al., 1990; Champigny et al., 1991; Popp and Gogelein, 1992; Ono et al., 1994, each of which is hereby incorporated by reference in its entirety. These non-astrocyte channels comprise a heterogeneous group with incompletely defined characteristics. They exhibit single-channel conductances in the range of 25-35 pS, discriminate poorly between Na+ and K+, are impermeable to anions, for the most part impermeable divalent cations, and they are blocked by similar concentrations of the adenine nucleotides ATP, ADP and AMP on the cytoplasmic side. The function of these non-selective ATP sensitive cation channels in these non-neural cell types remains enigmatic, in part because unphysiological concentrations of Ca2+ are generally required for channel activation.
  • Another subtype of ATP sensitive cation channel is the ATP-sensitive potassium channel (KAP channels) in pancreatic β cells. One class of insulin secretagogues, the antidiabetic sulfonylureas, are used to inhibit these KATP channels and stimulate insulin release in diabetes mellitus. See, Lebovitz, 1985. Antidiabetic sulfonylureas mediate their effect on KATP channels via a high affinity sulfonylurea receptor (SUR). See, Panten et. al., 1989; Aguilar-Bryan et. al., 1995. Several isoforms of the SUR, termed SUR1, SUWA, S W B, and SUR2c, have been identified and cloned. See, Aguilar-Bryan et. al., 1995; Inagaki et. al., 1996; Isomoto et. al., 1996; Lawson, 2000. These receptors belong to the ATP-binding cassette (ABC) transporter family, of which the cystic fibrosis transmembrane conductance regulator (CFTR), another ion channel modulator, is also a member. See, Higgins, 1992; Aguilar-Bryan et. al., 1995. Notably, the CFTR has major therapeutic importance, since its genetic absence causes cystic fibrosis, a fatal disease.
  • The sulfonylurea receptor imparts sensitivity to antidiabetic sulfonylureas such as glibenclamide and tolbutamide. Also, SUR is responsible for activation of the potassium channel by a chemically diverse group of agents termed K+ channel openers (SUR-activators), such as diazoxide, pinacidil, and cromakalin. See, Aguilar-Bryan et. al., 1995; Inagaki et. al., 1996; Isomoto et. al., 1996; Nichols et. al., 1996; Shyng et. al., 1997b. In various tissues, molecularly distinct SURs are coupled to distinct channel moieties to form different KATP channels with distinguishable physiological and pharmacological characteristics. The KATP channel in pancreatic β cells is formed from SUR1 linked with a K+ channel, whereas the cardiac and smooth muscle KATP channels are formed from SUR2A and SUR2B, respectively, linked to K+ channels. See, Fujita ad Kurachi, 2000.
  • Thus, a need exists for a physiological target instrumental in the cell swelling that is associated with brain damage in cerebral ischemia and traumatic brain injury and in the consequent morbidity and mortality. There is also a need for specific treatments for the cytotoxic edema that causes brain swelling, which worsens outcome and increases morbidity and mortality in brain injury and stroke. Also there exists a need for therapeutic compounds capable of modulating the activity of this target in order to prevent brain damage. The present invention is directed to a newly characterized non-selective calcium and ATP sensitive monovalent cation channel, termed the NCCa-ATP channel, which is present in neural cells and linked to an SUR. The present invention further provides a method to screen for or identify antagonists to NCCa-ATP channel activity. Further, the present invention provides a method for the therapeutic use of antagonists, such as sulfonylureas and other SUR1 blockers, to inhibit this channel's activity and thereby prevent neural cell swelling and cell death and the concomitant nervous system damage that includes brain swelling and brain damage.
  • SUMMARY OF THE INVENTION
  • The invention is based, in part, on the discovery of a specific channel, the NCCa-ATP channel, which is expressed in reactive neural cells after brain trauma. The present invention is directed to purified compositions containing a novel Ca2+-activated, [ATP]i, sensitive nonspecific cation channel, hereinafter the NCCa-ATP channel. In a preferred embodiment of the present invention, the compositions comprise mammalian neural cells or membrane preparations expressing the NCCa-ATP channel, most preferably the mammalian neural cells are freshly isolated reactive astrocytes. A preferred example of such a purified composition containing the NCCa-ATP channel is a membrane preparation derived from native reactive astrocytes. As demonstrated herein, when neural cells expressing the NCCa-ATP channel are depleted of intracellular ATP, the NCCa-ATP channel opens and the cells swell and die. However, if the NCCa-ATP channel is blocked on such cells, the cells do not swell and die. The invention is also based, in part, on the discovery that the NCCa-ATP channel is regulated by a type 1 sulfonylurea receptor, and that antagonists of this receptor are capable of blocking the NCCa-ATP channel and inhibit neural cell swelling.
  • The NCCa-ATP channel of the present invention is distinguished by certain functional characteristics, the combination of which distinguishes it from known ion channels. The characteristics that distinguish the NCCa-ATP channel of the present invention include, but are not necessarily limited to, the following: 1) it is a non-selective cation channels that readily allows passage of Na, K and other monovalent cations; 2) it is activated by an increase in intracellular calcium, and/or by a decrease in intracellular ATP; 3) it is regulated by sulfanylurea receptor type 1 (SURD, which heretofore had been considered to be associated exclusively with KATP channels such as those found in pancreatic β cells.
  • More specifically, the NCCa-ATP channel of the present invention has a single-channel conductance to potassium ion (K+) between 20 and 50 pS. The NCCa-ATP channel is also stimulated by Ca2+ on the cytoplasmic side of the cell membrane in a physiological concentration range, where said concentration range is from 10−8 to 10−5 M. The NCCa-ATP channel is also inhibited by cytoplasmic ATP in a non-physiological concentration range, where said concentration range is from 10−1 to 10 M. The NCCa-ATP channel is also permeable to the following cations; K+, Cs+, Li+, Na+; to the extent that the permeability ratio between any two of said cations is greater than 0.5 and less than 2.
  • The invention relates to assays designed to screen for compounds or compositions that modulate the NCCa-ATP channel, particularly compounds or compositions that act as antagonists of the channel, and thereby modulate neural cell swelling and the concomitant brain swelling. To this end, cell-based assays or non-cell based assays can be used to detect compounds that interact with, e.g., bind to, the outside (i.e., extracellular domain) of the NCCa-ATP channel and/or its associated SUR1. The cell-based assays have the advantage in that they can be used to identify compounds that affect NCCa-ATP channel biological activity (i.e., depolarization). The invention also provides a method of screening for and identifying antagonists of the NCCa-ATP channel, by contacting neural cells with a test compound and determining whether the test compound inhibits the activity of the NCCa-ATP channel. In one embodiment, methods for identifying compounds that are antagonists of the NCCa-ATP are provided. In one embodiment, therapeutic compounds of the present invention, including NCCa-ATP antagonists, are identified by the compound's ability to block the open channel or to prevent channel opening, by quantifying channel function using electrophysiological techniques to measure membrane current through the channel. NCCa-ATP antagonists include compounds that are NCCa-ATP channel inhibitors, NCCa-ATP channel blockers, SUR1 antagonists, SUR1 inhibitors, and/or a compounds that reduce the magnitude of membrane current through the channel. In this embodiment, channel function can be measured in a preparation of neural cells from a human or animal, and the test compound can be brought into contact with the cell preparation by washing it over the cell preparation in solution. The invention further provides a method of screening for sulfonylurea compounds that may act as antagonists of the NCCa-ATP channel.
  • The present invention relates to drug screening assays to identify compounds for the treatment of brain swelling, such as the swelling that occurs after brain injury or cerebral ischemia by using the NCCa-ATP channel as a target. The invention also relates to compounds that modulate neural cell swelling via the NCCa-ATP channel. The present invention also relates to the treatment of brain swelling by targeting the NCCa-ATP channel.
  • The invention also encompasses agonists and antagonists of the NCCa-ATP channel, including small molecules, large molecules, and antibodies, as well as nucleotide sequences that can be used to inhibit NCCa-ATP channel gene expression (e.g., antisense and ribozyme molecules). An antagonists of the NCCa-ATP channel includes compounds capable of (1) blocking the channel, (2) preventing channel opening, and/or (3) reducing the magnitude of membrane current through the channel.
  • The invention also encompasses the use of such compounds and compositions, that modulate NCCa-ATP channel activity to treat brain swelling. Further provided is a method of preventing brain swelling and the resulting brain damage through the therapeutic use of antagonists to the NCCa-ATP channel. In one embodiment, the therapeutic antagonist can be administered to or into the brain. Such administration to the brain includes injection directly into the brain, particularly in the case where the brain has been rendered accessible to injection due to trauma to the skull. The invention further provides the therapeutic use of sulfonylurea compounds as antagonists to the NCCa-ATP channel to prevent cell swelling in brain. In one embodiment the sulfonylurea compound is glibenclamide. In another embodiment, the sulfonylurea compound is tolbutamide.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 (comprised of FIGS. 1A, 1B, 1C, 1D, 1E and 1F); FIG. 1A shows whole cell current clamp recording before and after exposure to ouabain and before and after exposure to NaN3. FIG. 1B shows whole cell voltage-clamp recordings during ramp pulses (a) before and (b) after exposure to NaN3; (c) is the difference current. FIG. 1C shows whole cell voltage-clamp recordings during step pulses (a) before and (b) after exposure to NaN3; (c) is the difference current. FIG. 1D shows cell-attached patch recording of single ion channel openings induced by NaN3 at membrane potentials of (3) −80 mV and (4) 80 mV, compared to control patches at membrane potentials of (1) 80 mV and (2) −80 mV. FIG. 1E shows the cell-attached patch currents of FIG. 1D, shown at higher time resolution. FIG. 1F shows the cell-attached patch single-channel current-voltage relationship.
  • FIG. 2 (comprised of FIGS. 2A and 2B): FIG. 2A shows single channel currents recorded in an inside-out patch at different membrane potentials; dotted line indicates channel closing. FIG. 2B is a plot of inside-out patch single channel amplitude vs. membrane potentials.
  • FIG. 3 (comprised of FIGS. 3A, 3B, 3C and 3D); FIG. 3A shows single channel currents recorded in an inside-out patch with various alkaline ions substituting for K+ in the pipette; dotted line indicates channel closing. FIG. 3B is a plot of channel amplitude vs. membrane potential with various alkaline ions substituting for K+ in the pipette. FIG. 3C is a plot of channel amplitude measured in inside-out patches vs. voltage with Ca2+ and Mg2+ substituting for K+ in the pipette. To estimate channel pore size, FIG. 3D is a plot illustrating the relationship between the permeability (relative to Cs+) and the molecular radius of a series of monovalent organic cations, which included: (a) methanolmine, (b) guanidium, (c) ethanolamine, (d) diethylamine, (e) piperazine, (f) Tris, and (g) N-methylglucamine, data indicating an equivalent pore size of 0.67 nm.
  • FIG. 4 (comprised of FIGS. 4A and 4B); FIG. 4A shows single channel recordings in an inside-out patch in the absence and presence of cytoplasmic ATP. FIG. 4B is a plot of normalized open channel probability (n·Po) vs. concentration of cytoplasmic ATP.
  • FIG. 5. (comprised of FIGS. 5A and 5B); FIG. 5A shows current records from an inside-out patch exposed to different concentrations of [Ca2+]i. FIG. 5B the values of n·Po measured at the membrane potentials and [Ca2+]i indicated.
  • FIG. 6 is a plot of mean single channel amplitudes obtained in an inside-out patch configuration at different potentials studied and with different [Mg2+]i; the dotted line indicates 35 pS conductance.
  • FIG. 7 (comprised of FIGS. 7A and 7B) shows that presence of SUR1 mRNA and absences of Kir6.1 and Kir 6.2 in reactive astrocytes. Lanes 3 and 5 in FIG. 7A show the presence of SUR1 in insulinoma RIN-m5f cells and NRAs, respectively. Lanes 4 and 6 in FIG. 7A show that SUFU is absent in both cell types. Lanes 3 and 4 in FIG. 7B show that Kir6.1 is present in insulinoma RIN-m5f cells and Kir6.2 is absent from the insulinoma cells, respectively. Lanes 5 and 6 in FIG. 7B show that neither Kir6.1 nor Kir6.2 is present in NRAs, respectively.
  • FIG. 8 shows current recordings in an inside-out patch to illustrate the effects of tryptic digestion on channel sensitivity to glibenclamide and ATP.
  • FIG. 9 (comprised of FIGS. 9A and 9B) shows that the channel activator diazoxide can elicit channel activities under outside-out patch recording configuration. FIG. 9A shows the outside-out patch recordings with Na azide and diazoxide applied to the extracellular side of the membrane. FIG. 9B shows the current records obtained from the segments marked with the corresponding numbers in FIG. 9A, at higher temporal resolution.
  • FIG. 10 (comprised of FIGS. 10A, 10B and 10C) FIG. 10A shows outside-out patch recordings (a) before, (b) during, and (c) after application of glibenclamide to the extracellular side of the membrane. FIG. 10B shows the current records of FIG. 10A at higher temporal resolution. FIG. 10C show a plot of mean single channel amplitudes at the different potentials studied; the slope of the data indicates 35 pS conductance of the glibenclamide-sensitive channel.
  • FIG. 11 (comprised of FIGS. 11A and 11B) shows that sulfonylurea compounds inhibit channel activities. FIG. 11A shows the outside-out patch recordings with various concentrations of tolbutamide applied to the extracellular side of the membrane. FIG. 11B shows the dose-response curves for inhibition of open channel probability by glibenclamide and tolbutamide to provide a normalized open channel probability (n·Po); data were fit to a standard logistic equation, with a Hill coefficient of 1 and half-maximum inhibition of 48 nM and 16.1 μM; values plotted are means (± SE) from 3 and 5 patches for Glibenclamide and Tolbutamide, respectively.
  • FIG. 12 (comprised of FIGS. 12A, 12B, 12C, 12D, 12E, 12F, 12G, 12H and 12I); FIGS. 12A, 12B and 12C show the probability of channel opening in the presence of 0 μM, 3 μM, and 30 μM tolbutamide, respectively.
  • FIGS. 12D, 12E and 12F show the distribution of open channel dwell times in the presence of 0 μM, 3 μM, and 30 μM tolbutamide, respectively.
  • FIGS. 12G, 12H and 12I show the distribution of closed channel dwell times in the presence of 0 μM, 3 μM, and 30 μM tolbutamide, respectively.
  • FIG. 13 (comprised of FIGS. 13A, 13B and 13C) FIG. 13A shows outside-out patch recordings with diazoxide applied to the extracellular side of the membrane.
  • FIG. 13B shows current records at higher temporal resolution after application of diazoxide and at different membrane potentials.
  • FIG. 13C shows a plot of mean single channel amplitudes at the different potentials studied; the slope indicates 35 pS conductance of glibenclamide-sensitive channel.
  • FIGS. 14A, 14B and 14C are scanning electron micrographs of freshly isolated native reactive astrocytes. FIG. 14A shows the cells when formaldehyde-glutaraldehyde fixation was initiated under control conditions; FIG. 14B shows the cells fixed 5 min after exposure to 1 mM NaN3. FIG. 14C shows the cells fixed 25 min after exposure to 1 mM NaN3. Bar, 12 μm.
  • FIG. 15 (comprised of FIGS. 15A, 15B and 15C); FIG. 15A has photomicrographs of the epifluorescence images of cells exposed to different compounds and labeled with gropidium iodide (upper panel a, b and c) or annexin V (lower panel d, e and f). The compounds were: control (a & d), 1 mM Na azide (b & e), 1 mM Na azide plus 1 μM glibenclamide (c & f). FIG. 15B has bar graphs showing cell-counts for propidium iodide labeling; pairwise multiple comparisons indicated a significant difference (p<0.05) with Na azide treatment; FIG. 15C has bar graphs showing cell-counts for annexin V staining; pairwise multiple comparisons indicated no significant difference with any treatment.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
  • Some of the preferred embodiments of the present invention will be described in detail with reference to the attached drawings.
  • This invention may be embodied in many different forms and should not be construed as being limited to the embodiments set forth herein.
  • The present invention relates to a novel ion channel whose function underlies the swelling of mammalian neural cells, such as in response to ATP depletion; the use of the channel to screen for channel inhibitors, and the use of inhibitors of the channel function to prevent this cell swelling response, which characterizes brain damage in cerebral ischemia and traumatic brain injury.
  • Sodium azide (N a N 3) is a metabolic toxin used to induce “chemical hypoxia” by depleting intracellular ATP. See, Swanson, 1992. The morphological and electrophysiological responses of neural cells to NaN3 are examined in a novel cell preparation. Freshly isolated native reactive astrocytes (NRAs) from adult rat brain are used and studied in a native state immediately after their isolation. Reactive astrocytes are astrocytes that have been activated or stimulated in vivo, such as those associated with brain or neural injury. In the post-mortem brains of traumatic brain injury (TBI) patients, reactive astrocytes are found in proximity to the injury. The majority of reactive astrocytes surrounding an injury site in the brain are reactive astrocytes. Type 1 reactive astrocytes comprise >80% of recoverable reactive astrocytes, whereas type 2 reactive astrocytes comprise about 5%. Reactive astrocytes are normally polarized under quiescent conditions.
  • As used herein, the term “neural cells” includes astrocytes. The term “reactive astrocytes” means astrocytes found in brain at the site of a lesion or ischemia. The term “native reactive astrocytes” or WRAs” means reactive astrocytes that are freshly isolated from brain. The term “freshly isolated” as used herein refers to NRAs that have been purified from brain, particularly NRAs that were purified from about 0 to about 72 hours previously. When NRAs are referred to as being “purified from brain” the word “purified” means that the NRAs are isolated from other brain tissue and/or implanted gelatin or sponge and does not refer to a process that simply harvests a population of cells from brain without further isolation of the cells. As described herein, the NCCa-ATP channel found in reactive astrocytes is present only in freshly isolated cells; the NCCa-ATP channel is lost shortly after culturing the cells. NRAs provide an in vitro model that is more similar to reactive astrocytes as they exist in vivo in the brain, than astrocytes grown in culture. The terms “native” and “freshly isolated” are used synonymously. As used herein, the term “isolated neural cells” means neural cells isolated from brain.
  • Reactive astrocytes are produced in vivo and harvested from brain according to a method system similar to that described by Perillan. See, Perillan et al., 1999; Perillan et al., 2000. Harvested cells are then isolated and not cultured; rather, the freshly isolated reactive astrocytes are studied in a native state immediately after their isolation from the brain.
  • The Examples described herein reveal that NRAs from adult rat brain express a non-selective cation channel that is activated by depletion of [ATP]i at physiological concentrations of [Ca2+]i. This NCCa-ATP channel of the present invention, which is newly identified in NRAs and present in >90% of membrane patches from such cells, is distinguished from previously reported non-selective calcium and ATP channels by exhibiting significantly different properties. These distinguishing properties of the NCCa-ATP of the present invention include: being activated by submicromolar [Ca] and exhibiting a different sensitivity to block by various adenine nucleotides. Opening of the NCCa-ATP channel of the present invention by ATP depletion causes profound membrane depolarization, which precedes blebbing of the cell membrane. Upon ATP depletion, the NCCa-ATP channel opens to allow Na+ influx that leads to cell swelling. This channel is regulated by sulfonylurea receptor type 1 (SUR1). The channel can be blocked by sulfonylurea, such as glibenclamide and tolbutamide; treatment with glybenclamide results in significant reduction in swelling and blebbing induced by chemical ATP depletion. This channel participates in the cation flux involved in cell swelling. A method of the present invention includes the use of sulfonylurea compounds to inhibit the flow of current through the NCCa-ATP channel and inhibit blebbing related to channel opening. Also, use of sulfonylurea compounds and other compounds that inhibit the flow of current through the NCCa-ATP channel, thus can have a therapeutic preventative effect on cell swelling in brain.
  • Therefore, it is an object of the present invention to provide a composition comprising a membrane preparation expressing the NCCa-ATP channel. For example, the membrane preparation is derived from neural cells, such as isolated native reactive astrocytes (NRAs), preferably freshly isolated native reactive astrocytes. The NCCa-ATP channel in the composition has the following characteristics: (a) it is a 35 pS type channel; (b) it is stimulated by cytoplasmic Ca2+; (c) it opens when cytoplasmic ATP is less than about 0.804; and (d) it is permeable to the monovalent cations K+, Cs+, Li+ and Na+ and it can be blocked by antagonists of the type 1 sulfonylurea receptor.
  • Furthermore, it is an object of the present invention to provide a method of screening for antagonists of the NCCa-ATP channel, comprising: (a) contacting a test compound with a composition comprising the NCCa-ATP channel; and (b) identifying test compounds that inhibit an activity of said channel by measuring said activity in the presence and absence of said test compound, wherein a test compound that inhibits said activity is identified as an antagonist of the NCCa-ATP channel. For example, the composition may contain a preparation of neural cells expressing the NCCa-ATP channel or a membrane preparation expressing the NCCa-ATP channel, such as a membrane preparation derived from isolated native reactive astrocytes (NRAs). The effect of the compound on this channel may include: (a) blocking the NCCa-ATP channel; (b) closing the NCCa-ATP channel; (c) preventing the NCCa-ATP channel from opening; and (d) reducing the magnitude of membrane current through the NCCa-ATP channel. It is also an object of the present invention to identify a compound that is an NCCa-ATP antagonist, including an NCCa-ATP channel inhibitor, an NCCa-ATP channel blocker, a SUR1 antagonist, SUR1 inhibitor, and/or a compound capable of reducing the magnitude of membrane current though the channel.
  • It is a further object of the invention to provide a method for identifying compounds that inhibit neural cell swelling, comprising: (a) contacting a test compound with a composition comprising the NCCa-ATP channel, and (b) determining whether the test compound blocks the NCCa-ATP channel, wherein a test compound that blocks the NCCa-ATP channel is identified as a compound for inhibiting neural cell swelling.
  • It is a further object of the present invention to provide a method for identifying compounds that inhibit brain swelling, comprising: (a) contacting a test compound with a composition comprising the NCCa-ATP channel, and (b) determining whether the test compound blocks the NCCa-ATP channel, wherein a test compound that blocks the NCCa-ATP channel is identified as a compound for inhibiting brain swelling.
  • Yet another object of the present invention is to provide a method for identifying compounds that inhibit brain swelling, comprising: (a) contacting a test compound with a composition comprising the NCCa-ATP channel, and (b) determining whether the test compound inhibits neural cell swelling, wherein a test compound that inhibits neural cell swelling is identified as a compound for inhibiting brain swelling.
  • A further object of the present invention provides a method for identifying compounds that inhibit neural cell swelling in an animal, comprising: (a) contacting a test compound with a composition comprising the NCCa-ATP channel and determining whether the test compound blocks the channel, and (b) administering the test compound to an animal having a brain injury or cerebral ischemia, and determining whether the test compound that inhibits brain swelling of the treated animal, wherein test compounds that inhibit brain swelling are identified as compounds that inhibit neural cell swelling in an animal.
  • It is a further object of the present invention to provide a method for identifying compounds that inhibit brain swelling, comprising: (a) contacting a test compound with a composition comprising the NCCa-ATP channel, and determining whether the test compound blocks the channel, and (b) administering the test compound to an animal having a brain injury or cerebral ischemia, and determining whether the test compound inhibits brain swelling of the treated animal, wherein test compounds that block the NCCa-ATP channel are identified as compounds that inhibit brain swelling.
  • In each of these objects of the present invention, the composition preferably comprises a preparation of neural cells expressing the NCCa-ATP channel or a membrane preparation expressing the NCCa-ATP channel, which preferably is derived from isolated native reactive astrocytes (NRAs). It is a further object of the present invention to provide the above methods using a compound that is an antagonist of a type 1 sulfonylurea receptor, such as a sulfonylurea compound, a benzamido derivative or an imidazoline derivative.
  • It is a further object of the present invention to provide these methods in which the determining step include, but are not limited to, detecting or identifying swelling of the native reactive astrocytes, such as by microscopic observation of cell appearance (normal, blebbing, swelling); measuring channel currents; measuring membrane potential; detecting expression of annexin V; detecting expression of propidium iodide; in vitro binding assays; and combinations thereof.
  • It is a further object of the present invention to provide a method of preventing neural cell swelling in the brain of a subject, said method comprising administering to the subject a formulation containing an effective amount of a compound that blocks the NCCa-ATP channel and a pharmaceutically acceptable carrier.
  • It is a further object of the present invention to provide a method of alleviating the negative effects of traumatic brain injury or cerebral ischemia stemming from neural cell swelling in a subject, comprising administering to the subject a formulation comprising an effective amount of a compound that blocks the NCCa-ATP channel and a pharmaceutically acceptable carrier. Such administration may be delivery directly to the brain, intravenous, subcutaneous, intramuscular, intracutaneous, intragastric and oral administration. Examples of such compounds include antagonist of a type 1 sulfonylurea receptor, such as sulfonylureas like glibenclamide and tolbutamide, as well as other insulin secretagogues such as repaglinide, nateglinide, meglitinide, midaglizole, LY397364, LY389382, gliclazide, glimepiride, MgADP, and combinations thereof.
  • It is yet another object of the present invention to provide a formulation for preventing or inhibiting neural cell swelling in the brain of a subject, using a formulation that includes a compound that blocks the NCCa-ATP channel and a pharmaceutically acceptable carrier, wherein the quantity of said compound is at least 10 times greater than the quantity of said compound in formulations for treating diabetes.
  • EXAMPLES
  • The following examples are provided for further illustration of the present invention, and do not limit the invention. The examples provided herein are for illustrative purposes only, and are in no way intended to limit the scope of the present invention. While the invention has been described in detail, and with reference to specific embodiments thereof, it will be apparent to one with ordinary skill in the art that various changes and modifications can be made therein without departing from the spirit and scope thereof. Experiments and exemplary procedures are described below which provide additional enabling support for the present invention. In particular, in vitro studies using freshly isolated reactive astrocytes and in vivo studies using appropriate animal models are disclosed.
  • General Methods Cell Preparation
  • Reactive astrocytes are produced in vivo and harvested from adult brain in the following manner: gelatin sponges (Gelfoam®, Upjohn Co., Kalamazoo Mich.) are implanted into a stab wound in the parietal lobe of 8 week old Wistar rats as described herein. Sponge pieces are harvested at 8 days and washed three times in phosphate-buffered saline (PBS, pH 7.4) to remove adherent tissue. Depending on the number of NRAs required for a particular study, the sponge pieces may be harvested earlier or later after implantation into a stab wound, with the preferred harvest being conducted from about 2 days to about 30 days after implantation, and the most preferred range being conducted from about 2 days to about 3 days after implantation.
  • NRAs are freshly isolated from the sponge pieces in the following manner: washed pieces are placed in an Eppendorf tube containing artificial cerebrospinal fluid (aCSF) composed of (mM): 124 mM NaCl, 5.0 mM, 1.3 mM MgCl2, 2.0 mM CaCl2, 26 mM NaHCO3, and 10 mM D-glucose; at pH 7.4, ≈290 mOsm, wherein the aCSF contains papain 20 U/ml, trypsin inhibitor 10 mg/ml and DNase 0.01% (Worthington, Lakewood, N.J.), the entirety of which is referred to as a “digestion system.”
  • This digestion system is transferred to an incubator (humidified 90%/10% air/C02, 37° C.) for 20 minutes, and is gently triturated every 5 minutes. The cell suspension is centrifuged at 3,000 rpm for 1 minute. The pelleted cells are resuspended in aCSF and stored at 4° C. until studied.
  • For some studies, prior to resuspension in aCSF, the pelleted cells can be further purified by removing red blood cells (RBCs) using density gradient centrifugation in Histopaque-1 077 (Sigma Diagnostics, St. Louis, Mo.). This further purification process can produce a population of cells in which <<1% are RBCs, as determined by phase contrast microscopy.
  • Scanning Electron Microscopy (SEM)
  • To study cell blebbing and swelling, freshly isolated cells are exposed at room temperature to NaN3 then, after various time intervals, cells are fixed using iced 4% formaldehyde+1% glutaraldehyde for 24 hours then dehydrated using serial concentrations (35, 50, 75, 95, 100%) of ethanol. Specimens are critical point dried (Tousimis), gold coated (Technics), and viewed using an AMR 1000 scanning electron microscope.
  • Electrophysiology
  • Experiments are carried out at room temperature, 22-25° C., using NRAs within 24 hour of cell isolation. An aliquot of these freshly isolated NRAs is placed in the recording chamber filled with extracellular bath solution containing (a): NaCl 130, KCl 10, CaCl 2 1, MgCl 2 1, HEPES 32.5, glucose 12.5, pH 7.4. After viable cells adhere to the surface, flushing with excess solution washes away residual debris not previously removed by centrifugation. Membrane currents are amplified (Axopatch 200A, Axon Instruments, Foster City, Calif.) and sampled on-line at 5 kHz using a microcomputer equipped with a digitizing board (Digidata 1200A, Axon Instruments) and running Clampex software (version 8.0, Axon Instruments). Membrane currents are recorded in intact cells using both the cell-attached and the nystatin-perforated whole-cell configurations, according to methods described in Horn and Marty, 1988. Membrane currents are recorded in cell-free isolated membrane patches, using both the inside-out and outside-out configurations, such as those described in Hamill et al., 1981. Patch clamp pipettes, pulled from borosilicate glass (Kimax, Fisher Scientific, Pittsburgh, Pa.), have resistances of 6-8 MΩ a for single channel recordings and 2-4 MΩ a for experiments using the nystatin-perforated whole-cell technique. The bath electrode is a Ag/AgCl pellet (Clark Electromedical, Reading, England) that is placed directly in the bath except when the bath [Cl−] is altered, in which case an agar bridge made with 3 M KCl is used to connect to the bath.
  • The terms “intracellular” and “cytoplasmic” are interchangeable, as are the terms “extracellular” and “external”. The terms “voltage” and “potential” are interchangeable when referring to membrane voltage or membrane potential. “Clamping” a cell membrane refers to holding the voltage across the cell membrane constant and measuring changes in membrane current as membrane resistance changes due to ion channel opening and closing (“voltage clamp”) or holding the current across the cell membrane constant and measuring changes in membrane voltage as membrane resistance changes due to ion channel opening and closing (“current clamp”). When a membrane voltage is imposed on the cell, for example with a “ramp” or “pulse”, it is understood that the cell membrane has been voltage-clamped and membrane current is being measured. When membrane “resting potential” is measured, it is understood that the cell membrane has been current-clamped and membrane voltage is being measured.
  • The “whole-cell” experimental configuration refers to a situation in which a recording pipette penetrates the cell membrane so that the pipette solution is continuous with the cytoplasm or the membrane under the pipette is perforated using nystatin, the external solution is in contact with the extracellular membrane, and current or voltage recordings represent measurements from the entire cell membrane. The “cell-attached patch” experimental configuration refers to a situation in which the pipette contacts the cell so that the patch is still forming part of the intact cell membrane and channels in the patch are recorded. The “outside-out patch” experimental configuration refers to a situation in which an excised patch of cell membrane is sealed to the tip of a recording pipette so that the pipette solution is in contact with the extracellular side of the membrane, the external solution is in contact with the cytoplasmic side of the membrane, and current or voltage recordings represent measurements from the excised patch of membrane. The “inside-out patch” experimental configuration refers to a situation in which an excised patch of cell membrane is sealed to the tip of a recording pipette so that the pipette solution is in contact with the cytoplasmic side of the membrane, the external solution is in contact with the extracellular side of the membrane, and current or voltage recordings represent measurements from the excised patch of membrane.
  • The term “patches” includes, but is not limited to: inside-out patches, outside-out patches, an excised patch of a cell membrane, or a cell-attached patch. The term “membrane preparation” includes patches as well as cell membranes isolated from mammalian cells or tissues. Isolated mammalian cell membranes are produced by methods well known in the art. One example of such a membrane preparation is a microsomal fraction purified from disrupted cells or a tissue sample by discontinuous sucrose gradient centrifugation.
  • Patches with seal resistance of <3 GΩ and access resistance of >50 MΩ are discarded. Macroscopic membrane currents are measured during step pulses (600 ms) or during ramp pulses (−140 to +50 mV at 0.32 mV/ms) from a holding potential of −67 mV.
  • Recording Solutions
  • For whole cell macroscopic recordings, a nystatin perforated patch technique is used, with a bath solution containing (mM): NaCl 130, KCl 10, CaCl 2 1, MgCl 2 1, HEPES 32.5, glucose 12.5, pH 7.4. The pipette solution contains (mM): KCl 55, K2SO4 75, MgCl 2 8, and HEPES 10, pH 7.2. Nystatin, 50 mg (Calbiochem) is dissolved in dimethylsulfoxide (DMSO), 1 ml. Working solutions are made before the experiment by adding 16.5 μl nystatin stock solution to 5 ml of the base pipette solution to yield a final concentration of nystatin of 165 μg/ml and DMSO 3.3 μl/ml. This composition of the pipette solution includes K2SO4 instead of a portion of the KCl that would otherwise be included. The S042-anion, unlike Cl−, is not permeable through the nystatin pore. Reducing the pipette [Cl−] reduces the driving force for Cl− into the cell, thereby minimizing osmotic swelling of the cell that might otherwise occur during electrophysiological recording (Horn and Marty, 1988).
  • For cell-attached patch recording, a bath solution is used containing (mM): NaCl 130, KCl 10, CaCl 2 1, MgCl 2 1, HEPES 32.5, glucose 12.5, pH 7.4. The pipette contains (mM): KCl 145, MgCl 2 1, CaCl2 0.2, EGTA 5, HEPES 10, pH 7.28. The measured osmolarity of the extracellular solution is ≈300 mOsm (Precision Systems, Natick, Mass.).
  • For most inside-out patch recording, a bath solution is used containing (mM): CsCl 145, CaCl2 4.5, MgCl 2 1, EGTA 5, HEPES 32.5, glucose 12.5, pH 7.4. The pipette contains (a): CsCl 145, MgCl 2 1, CaCl2 0.2, EGTA 5, HEPES 10, pH 7.28. For other inside-out patch recordings, Cs+ in the above solutions is replaced with equimolar K+.
  • For the inorganic cation substitution experiments, Cs+ in the pipette is replaced by equimolar concentrations of individual test ions, except when using Ca2+ or Mg2+, in which cases a concentration of 75 mh4 is used to facilitate seal formation (Cook et al., 1990).
  • For outside-out patch recording, the pipette solution contains (mM): CsCl 145, MgCl 2 1, CaCl2 0.2, EGTA 5, HEPES 10, pH 7.28. The standard bath solution contains (mM): CsCl 145, CaCl24.5, MgCl 2 1, EGTA 5, HEPES 32.5, glucose 12.5, pH 7.4. For the organic cation substitution experiments, Cs+ in the bath is replaced with equimolar concentrations of test cation.
  • For experiments requiring low concentration of free Ca2+ in bath solution, Ca2+-EGTA buffered solution is employed, and free [Ca2+] is calculated using the program WEBMAXC v2.10 (http://www.stanford.edu/-cpatton/maxc.html). For [Ca2+]=1 μM, 5 mM EGTA is used and 4.5 mM Ca2+ salt. [Ca2+]=1 μM is also used in solutions to test intracellular ATP and Mg2+ activities.
  • Single-channel amplitudes used to calculate slope conductance are obtained by fitting a Gaussian function to an all-points amplitude histogram of records obtained at various potentials. To calculate open channel probability (n·Po) at various potentials and with different test agents, the all-points histogram is fit to a Gaussian function and the area under the fitted curve for the open channel is divided by the area under the fitted curve for the closed plus open channel. Values of n·Po at different concentration of test agents are fit to a standard logistic equation using a least-squares method.
  • For estimating ionic permeabilities of various cations relative to that for K+, each permeability (Px/PK) is obtained from its reversal potential (Erev) by fitting to the Goldman-Hodgkin-Katz (GHK) equation well known in the art. See Goldman 1943; Hodgkin and Katz, 1949. Current-voltage data are fit to the GHK equation, assuming that both K+ and the test ion are permeant.
  • To estimate the pore size of the NCCa-ATP channel of the present invention, the relative permeabilities of organic cations are evaluated. The Stokes-Einstein radius (rse) is calculated from the limiting conductivities (*) of the ions with the formula: rSE·λ=constant. The constant is determined from the behavior of TEA at 25° C., for which λ=44.9 cm2Ω-1, rSE=0.204 nm. The Stoke-Einstein radius is then converted to the molecular radius using correction factors read off from Figure 6.1 in Robinson and Stokes, 1970. The equivalent limiting conductance for ethanolamine is given (ibid.) and those of other ions are calculated from their molecular weight by the formula, MW 0.5·λ=constant. The constant is determined by the value for ethanolamine at 25° C.: MW=62.1 and λ=4.42 cm2Ω-1 equiv. Relative permeabilities (Px/PCs) are then plotted against the calculated ionic radii. The effect of solute size on the rate of penetration (permeability) through pores is expressed by the Renkin equation (Renkin, 1955):

  • a/a 0=[1−(r/R)][1−2.104(r/R)+2.09(rR)3−0.95(r/R)5]  (1)
  • in which a, a0, r, and R are the effective area of the pore, the total cross sectional area of the pore, radius of the solute, and radius of the pore, respectively.
  • Junction potentials are determined with an electrometer by measuring the diffusion potential established across a dialysis membrane and are subtracted when appropriate. Holding currents are not subtracted from any of the recordings. Difference currents are obtained by simply subtracting current records before and after perfusing NaN3, with no other processing being employed.
  • Results
  • The Role of the NCCa-ATP Channel in Neural Cell Swelling
  • Characterization of the Channel
  • Example 1 Morphological Changes with ATP Depletion Using NaN3
  • Cultured neural cells have been shown to swell upon ATP depletion. See, Jurkowitz-Alexander et al., 1992; Jurkowitz-Alexander et al., 1993. Freshly isolated NRAs depleted of ATP also results in cell swelling. Ischemia or traumatic injury in brain also causes depletion of ATP in brain neural cells.
  • The surfaces of freshly isolated NRAs are highly complex, exhibiting small membrane evaginations and fine processes that decorate the entire cell surface, as shown in the scanning electron micrograph in FIG. 14A. Exposure of NRAs to NaN3 (1 mM) causes changes in the surface appearance, characterized early-on by loss of complex structure and development of surface blebs (FIG. 14B), followed later by a grossly swollen appearance with complete loss of fine structure and formation of multiple large blebs (FIG. 14C). Therefore, NRAs undergo blebbing and swelling after NaN3-induced ATP depletion.
  • Phase contrast microscopy is also useful for assessing this process, although fine structure cannot be resolved. Blebbing is visibly apparent 10-15 minutes after exposure to NaN3. Morphological changes of this sort are attributable to loss of cytoskeletal integrity, combined with action of an osmotic force that causes swelling of the cell.
  • To assess the contribution of the osmotic gradient to cell swelling, the experiment is repeated in the presence of mannitol, an impermeant oncotic agent. Mannitol (50 mM), at a concentration sufficient to increase osmolarity of the extracellular solution from 300 to 350 mOsm, delays bleb formation>30 minutes after exposure to NaN3. Cellular ATP also can be depleted using exposure to NaCN (2.5 mM) plus 2-deoxyglucose (10 mM). See, Johnson et al., 1994. Similar morphologic changes, including cell membrane blebbing and delay of blebbing by mannitol are obtained following exposure to NaCN and 2-deoxyglucose. This demonstrates that the effect of NaN3 is due in fact to ATP depletion and not to any other non-specific effect of NaN3.
  • Example 2 General Electrophysiological Properties of NRAs
  • The macroscopic currents of whole cell preparations of N u s are characterized by small inward currents at negative potentials, large outward currents at positive potentials, and a flat “plateau” region at intermediate potentials. NRAs exhibit macroscopic currents that are consistent with observations in primary cultured cells of the same origin. See, Perillan et al., 1999; Perillan et al., 2000. The NRAs exhibited inward currents negative to the K+ equilibrium potential (EK) are usually <100 pA, much smaller than values reported in cultured neonatal astrocytes (Ransom and Sontheimer, 1995), but consistent with findings in astrocytes freshly isolated from injured brain (Bordey and Sontheimer, 1998; Schroder et al., 1999). The large outward currents in NRAs are partially blocked by charybdotoxin (100 nM), iberiotoxin (1 00 nM) and tetraethylammonium chloride (5 mM), consistent with the presence of a large conductance Ca2+-activated K+ channel. See, Perillan et al., 1999. The outward current that remains in the presence of charybdotoxin can be further blocked by 4-aminopyridine (5 mM), and exhibits kinetic properties typical of a delayed rectifier K+ channel. Consistent with a previous report (Perillan et al., 1999), fast inward voltage dependent currents attributable to Na+ channels are observed in less that 1% of NRAS.
  • NaN3 Elicits Depolarizing Inward Current Due to 35 DS Channel
  • Current clamp recordings are used to investigate the effect of ATP depletion by NaN3 in NRAs. For these experiments, a nystatin-perforated patch method is used to assure that the metabolic disruption comes from drug application and not cell dialysis. Extracellular application of NaN3 (1 mM; room temperature) results in a large and swift depolarization of the cells (FIG. 1A). NaN3 rapidly depolarizes the cells to Em≈0 mV (−4.3±0.9 mv). Depolarization usually starts −1 minute after addition of NaN3, is complete in <3 minutes, and is irreversible on washout of drug. Ouabain is a known Na+/K+-ATPase blocker. See, Brismar and Collins, 1993. The magnitude of the depolarization observed with NaN3 far exceeds the small reversible depolarization induced by ouabain (1 mM). This indicates that the large depolarization observed after exposure to NaN3 is not caused by Na+/K+-ATPase pump failure.
  • The time course of depolarization with NaN3 is appreciably more rapid than the time course for development of cell membrane blebbing observed with the same treatment. Also, neither the time course nor the magnitude of the depolarization is affected by raising the extracellular osmolarity with 50 mM mannitol, a treatment that substantially delays bleb formation. Thus, depolarization is a primary event, not secondary to cell swelling or stretch.
  • Voltage-clamp recordings show that exposure to NaN3 results in a net increase of inward current in NRAs. Recordings obtained using both ramp (FIG. 1B) and step pulses (FIG. 1 C) show significantly larger currents after NaN3 treatment, as shown by comparing the recordings before (a) and after (b) NaN3 treatment. A plot of the “difference currents”, obtained by subtracting the current-voltage curve before drug from that after drug (line c in FIG. 1B), indicates that the new current turned on by NaN3 reverses near 0 mV. A reversal potential near 0 mV is indicative that the NaN3-induced current results from a non-specific cation conductance.
  • To further characterize the NaN3-induced current, cell-attached patch recordings are used. Exposure to NaN3 elicits single channel currents in patches that exhibit no single channel currents prior to addition of drug (FIG. 1D). After addition of NaN3, recordings at low temporal resolution reveal a large increase in current variance that, after increasing temporal resolution, is revealed to be due to single channel events (FIG. 1E at 3 and 4). The amplitudes of single-channel events recorded at different membrane potentials are plotted in FIG. 1F, which shows that NaN3 activates a single channel conductance of ≈35 pS that exhibits weak inward rectification when measured in the cell-attached configuration.
  • Additional experiments are carried out in the cell-attached configuration with the pipette solution supplemented with various drugs. The NaN3-induced single channel currents are not blocked by 10 mM TEA, 5 mM 4-AP, 100 nM iberiotoxin, 100 nM charybdotoxin, or 1 μM tetrodotoxin (4-6 patches for each compound). These experiments indicating that a typical K+ or Na+ channel is not involved. Also, because 0.2 mM Ca2+ is included in the pipette solution, these single channel openings are unlikely to be due to monovalent cation influx via an L-type Ca2+ channel.
  • Similar depolarization and activation of a 35 pS channel are obtained when cellular ATP is depleted using exposure to NaCN (2.5 mM) plus 2-deoxyglucose (10 mM). This demonstrates that the effect of NaN3 is caused by ATP depletion and not by any other non-specific effect of NaN3.
  • Apart from ATP depletion, patch excision is also a highly reliable method for channel activation. Of the more than 120 cells studied in the cell-attached configuration, spontaneous channel activity attributable to a ≈35 pS conductance is detected in only 2 cells. Thus, the NCCa-ATP channel of the present invention is typically silent in metabolically healthy cells. By contrast, a ≈35-pS channel is present in >90% of inside-out patches formed from NRAs not exposed to NaN3 or other metabolic toxins, thus demonstrating that an intracellular element lost on patch excision normally prevents channel activation.
  • Another potential mechanism of channel activation other than patch excision is regulatory volume decrease (RVD). Cell swelling is widely recognized as a stimulus that initiates RVD, a phenomenon accompanied by activation of various currents, including a non-selective cation channel in some systems. See, Ono et al., 1994. When membrane patches are studied in a cell-attached configuration, hyposmotic stimulation (210 mosmo/kgH2O) activated single channel events, but none exhibit a ≈35 pS conductance. This finding indicates that the depolarization and channel activation observed with NaN3 are not part of an RVD response secondary to NaN3-induced cell swelling, and accords with the previously noted observation that NaN3-induced depolarization preceded cell swelling. This fact is supported by the observation that the NCCa-ATP channel is seldom observed in cell attached patches from healthy cells, but becomes evident in >90% of patches after conversion to an inside-out configuration. Also, the NCCa-ATP channel is lost shortly after culturing reactive astrocytes.
  • Example 3 Relative Permeabilities and Pore-Size
  • The channel is further characterized using membrane patches in the inside-out configuration. Records obtained during test pulses to various potentials with equal [K+] on both sides of the membrane are shown in FIG. 2A. Amplitude histograms are constructed of events observed at potentials from −140 mV to +100 mV, and values (mean±SE) for 4 patches are plotted and show in FIG. 2B. Fit of the data to a linear equation indicates a slope conductance of 35 pS, with an extrapolated reversal potential (Erev) of +0.1 mV, close to the expected K+ reversal potential (EK) of 0 mV.
  • In addition to conducting K+, the channel transports a variety of alkaline ions (FIG. 3A), indicating that it is a non-selective cation channel. In inside-out patches, the conductance of the channel is measured with various alkaline ions in the pipette solution, including Cs+, Na+, Rb+, K+, and Li+, always with equimolar K+ in the bath solution. Current-voltage data are fit to the GHK equation. Na+ is shown to have a nearly equal slope conductance (32.6 pS) compared to K+ (35.2 pS), but the slope conductance is reduced with other cations (FIG. 3B). Measurements of Erev are used to estimate relative permeabilities for the series of alkaline ions. Values for relative permeabilities derived from the GHK equation are PCs +/PK +=1.06, PNa +/PK +=1.04, PRb +/PK +=1.02, and PLi +/PK+=0.96, indicating that this channel is nearly equally permeable to all monovalent cations.
  • The permeability of the NCCa-ATP channel of the present invention to anions, such as Cl, is also assessed. After measuring single channel current amplitudes at different potentials with 145 mM KCl, the bath solution is changed to equimolar K+ gluconate. When an agar bridge is used, the solution change resulted in a change in Erev<0.5 mV, indicating that the NCCa-ATP channel of the present invention is essentially impermeable to anions.
  • The permeability of the instant channel to divalent cations, Ca2+ and Mg2+, is also investigated (FIG. 3C). When potassium ion in the pipette solution is replaced with 75 mM Ca2+ or Mg2+, inward currents are not detected. Fit to the GHK equation gives best fit values for Erev,<<−65 mV for Ca2+ and Mg2+ respectively, giving relative permeabilities with respect to K+ of <<0.001, indicating that this channel is essentially impermeable to divalent cations.
  • Because the NCCa-ATP channel of the present invention discriminates very poorly among monovalent inorganic cations (FIGS. 3A and B), experiments are performed to determine the equivalent pore size of the channel by measuring channel permeability, relative to Cs+, for a wide range of organic cations. Using an outside-out patch configuration, single-channel current-voltage relations are plotted to obtain Erev for a number of organic cations. Permeability ratios are then derived from fits to the GHK equation. For each of the organic cations (a) nethanolamine, (b) guanidium, (c) ethanolamine, (d) diethylamine, (e) piperazine, (f) Tris, and (g) N-methylglucamine, the mean value of relative permeability measured is plotted against its hydrated molecular radius (FIG. 3D, empty circles). The permeability ratios define a smoothly declining series of values that are well fit by the Renkin equation. The Renkin equation describes the permeation of a rigid sphere through a cylindrical pore. Renkin, 1955. Least-squares, fit to the equation, indicates an equivalent pore radius of 0.67 nm for the NCCa-ATP channel of the present invention. A 0.67 nm pore radius is similar to pore sizes of 6 A, found for the Ca2+ channel (McCleskey and Almers, 1985) and 7.4 A, found for the nAChR channel (Adams et al., 1980). Junction potentials determined according to the methods described herein generally did not exceed 5 mV.
  • Example 4 Inhibition by [ATP]i
  • The NCCa-ATP channel is inhibited by intracellular ATP, based on the finding that this channel is turned on after depleting intracellular ATP by exposure to NaN3 (See FIGS. 1B, 1C, 1D and 1E) or to NaCN plus 2-deoxyglucose. This fact is supported by the observation that the NCCa-ATP channel of the present invention is seldom observed in cell attached patches from healthy cells, but becomes evident in >90% of patches after conversion to an inside-out configuration.
  • Inside-out patches are used to demonstrate that the channel is sensitive to block by ATP on the cytoplasmic side of the membrane. Patches are studied using Cs′ as the charge carrier, to assure that no K+ channel, such as Kir2.3 or KATP, is contributing to patch activity. With no ATP and 1 μM Ca2+ in the bath, the NCCa-ATP channel exhibits vigorous openings. 1 mM ATP causes profound diminution in channel activity, an effect that is readily reversed on washout (FIG. 4A); however, channel availability is unaffected by 1 mM AMP or ADP. The open channel probability (n·Po) is measured at different [ATPIi, and these values are normalized to that obtained at [ATP]i=O mM, and fitted to a standard logistic equation. As shown in FIG. 4B, the NCCa-ATP channel is blocked by [ATP], in a dose-dependent manner. Half maximum inhibition (IC50) is observed at [ATP]i, =0.79 μM with a Hill coefficient of 1, and channel activity is completely abolished at [ATP]i>30 μM. ADP and AMP, have no effect on the NCCa-ATP channel activity in inside-out patches.
  • This in vitro assay for determining the concentration of the test compound which achieves a half-maximal inhibition of channel activity (ICSO) may be used to formulate dose in animal models to achieve a circulating plasma concentration range that includes the IC50.
  • Example 5 Activation by [Ca2+]i
  • The Ca2+ concentration on the cytoplasmic side of the membrane is also found to regulate activity of the NCCa-ATP channel of the present invention. The relationship between NCCa-ATP channel activity and [Ca2+]i is examined using inside-out patches studied at membrane potential (Em)=−80 mV. Changing [Ca2+]i clearly affects activity of the NCCa-ATP channel (FIG. 5A). When free [Ca2+]i is <30 nM, no channel activity is apparent. With [Ca2+]i>30 nM, the open probability (no·o) increases in accordance with the [Ca2+]i, up to ≈1 μM of [Ca2+]i at which activity is near maximum. The effect of Ca2+ on channel availability is found to depend on membrane voltage. Values of no·o from 4-9 patches obtained at three different potentials, Em=−40 mV, −80 mV and −120 mV, are normalized to values observed with 3 μM [Ca2+]i. These data are fit to a standard logistic equation using a Hill coefficient of 1.5 and half-maximum values of 0.12 μM, 0.31 μM and 1.5 μM at −40 mV, −80 mV and −120 mV, respectively (FIG. 5B). These data indicate that channel activity is strongly dependent on [Ca2+]i at physiologically relevant concentrations, and that the effect of Ca2+ is voltage dependent, consistent with a Ca2+ binding site inside the electric field of the membrane.
  • Example 6 Internal Mg2+ Causes Rectification
  • Because certain channels are sensitive to intracellular Mg2+ (Chuang et al., 1997; Perillan et al., 2000), experiments are carried out to determine whether the channel rectification observed in cell-attached patch recordings (see FIG. 1 F) might be due to intracellular Mg2+. Using inside-out patches studied with equimolar K+ on both sides of the membrane, [Mg2+] is varied on the cytoplasmic side. Single channel records and channel amplitudes observed with different [Mg2+]i are shown (FIG. 6). No rectification is evident with [Mg2+]i 30 μM, but at [Mg2+]i≈100 μM, increasingly strong rectification is present. At 100 μM, Mg2+ appears to produce a flickery block.
  • Example 7 Identifying the Presence of SUR in NRAs
  • To determine if SUR receptors are present in NRAs, the binding of glibenclamide to these cells is assessed by fluorescence microscopy. Eight week old Wistar rats are injured by a stab wound into the subcortical white matter and implantation of a gelatin sponge as previously described herein. Eight days later, tissue sections of formaldehyde-fixed brains from injured animals are incubated for 60 minutes at room temperature with 20 nM FITC-conjugated glibenclamide. A fluorescence image of the gelatin sponge shows labeled cells lining the cavities of the sponge. In brain adjacent to the injury, essentially no glibenclamide binding is apparent. These data indicate that SUR, which are not normally present in subcortical white matter, are expressed in neural cells following traumatic injury.
  • RT-PCR
  • Total RNA is extracted from cells and used to synthesize cDNA, which is amplified from reactive astrocytes is analyzed by RT-PCR on an agarose gel stained with ethidium bromide. FIG. 7A is a photograph of the gel showing the RT-PCR for SUR1 and SUR2. FIG. 7B is a photograph of a gel showing the RT-PCR for Kir6.1 and Kir6.2. Lanes 3 and 4 in FIGS. 7A and 7B show the RT-PCR for insulinoma cells. Lanes 5 and 6 show the RT-PCR for reactive astrocytes. Lane 1 in FIGS. 7A and 7B represents ladder size markers; Lane 2 in FIGS. 7A and 7B is a blank control. In FIG. 7A, lanes 3 and 4 show the SUR1 and SUR2 experiments, respectively, in insulinoma cells. Insulinoma cells are known to express SUR1, but not SUR2. Lanes 5 and 6 in FIG. 7A show the SUR1 and SUR2 experiments in reactive astrocytes, respectively. FIG. 7A shows that SUR1 mRNA is present in reactive astrocytes, as well as in the control insulinoma cells. SUR2 is absent in both cell types. In FIG. 7B, lanes 3 and 4 show the Kir6.1 and Kir6.2 experiments in insulinoma cells, respectively. Kir6.1 is present in insulinoma cells, but Kir6.2 is not. Kir6 is the potassium channel associated with SUR1 in insulinoma cells. Lane 5 and 6 in FIG. 7B show that neither Kir6.1 nor Kir6.2 is present in reactive astrocytes. Therefore, reactive astrocytes express SUR1 mRNA, but Kir6.1 and Kir6.2 mRNA is absent from the cells.
  • The presence of SUR1 in reactive astrocytes combined with the regulation of the NCCa-ATP channel in astrocytes by SUR antagonists indicates that SUR regulates the NCCa-ATP channel of the present invention.
  • Example 8 Tryptic Digests
  • A characteristic feature of SUR-regulated K A˜function is that tryptic digestion of the cytoplasmic face of the channel, but not its extracellular face causes loss of inhibition by sulfonylureas, without altering sensitivity to ATP and without changing the biophysical properties of the channel. The effect of trypsin on NCCa-ATP function is shown in FIG. 8. Under control conditions, channel activity in the inside-out patch configuration is strongly inhibited by 1 pM glibenclamide. Exposure to 100 pg/ml trypsin on the cytoplasmic side of the membrane for 3 minutes yields a patch that still exhibits strong channel activity, but that channel activity is completely unaffected by glibenclamide. After such trypsin treatment of the cytoplasmic side, the biophysical properties of the channel, including open channel conductance, open channel times, Ca2+-mediated activation are unchanged, and the channel still maintains its typical sensitivity to ATP. By contrast, exposure of the extracellular side of the membrane has no effect on glibenclamide inhibition. These trypsin digest data on the NCCa-An channel of the present invention provide additional supporting evidence that SUR1 is involved in regulation of the NCCa-ATP channel, because the results compare to previous findings from SUR1-regulated KATP channels. Linkage of a SUR to a non-selective ATP sensitive cation channel, has not been shown previously.
  • Assays for Compounds or Compositions that Block NCCa-ATP Channel and Inhibit Neural Cell Swelling
  • Example 9 Effects of Sulfonylurea Compounds
  • Sulfonylurea compounds are known to modulate the sulfonylurea receptor. A sulfonylurea receptor is generally associated with KATP channels as a regulatory component, and is found in various tissues, including rat NRAs. Notably, the KATP channels Kir6.1 and Kir6.2 are not present in rat NRAs (FIG. 7B). It is possible to activate the NCCa-ATP channel with SUR ligand diazoxide in outside-out patches (FIGS. 9A and 9B). NaN3 does not elicit channel activity in isolated membrane patches, indicating that it works via ATP depletion rather than any direct effect on the channel.
  • Example 10 In Vitro Assays for Determining Dose-Dependent Blockage of the NCCa-ATP Channel
  • SUR1 blocking compounds, such as glibenclamide and tolbutamide, are known to have an inhibitory effect on KATP channels. h one embodiment, the present invention arrives at the objects of the invention by providing a method in which the direct inhibitory effect of glibenclamide and tolbutamide on NCCa-ATP channels is determined (FIGS. 10 and 11). Inside-out patches are used to show the inhibitory effect of sulfonylureas. To ensure that no K+ channel, particularly KATP is contributing to patch current, Cs+ is used as the charge carrier. Channel activity is profoundly diminished by the addition of 10 μM glibenclamide (FIGS. 10A at b), and the activity is shown to be due to a 35 pS cation channel, which is consistent with the NCCa-ATP channel of the present invention (FIG. 10C). Another sulfonylurea, tolbutamide, is also shown to inhibit NCCa-ATP channel activity (FIGS. 11A and 11B). AS shown in FIG. 11 B, the NCCa-ATP channel is blocked by the sulfonylureas in a dose-dependent manner. With tolbutamide, half maximum inhibition (EC50) is observed at 16.1 μM with a Hill coefficient of 1.3, and channel activity is completely lost at concentrations>300 μM. With glibenclamide, EC50 is observed at 48 μM with a Hill coefficient of 1.2. The sensitivity of the NCCa-ATP channel of the present invention to blocking in NRAs with both of these sulfonylurea compounds corresponds closely to that reported in pancreatic p cells and in expression systems with SUR1, but not SUE.
  • This in vitro assay for determining the concentration of the test compound which achieves a half-maximal inhibition of channel activity may be used to formulate dose in animal models to achieve. a circulating plasma concentration range.
  • Example 11 Mechanism of Channel Regulation by Sulfonylureas
  • The NCCa-ATP channel of the present invention exhibits two open states, with a shorter and a longer dwell time, each less than 10 ms. FIG. 12 shows data from a patch exhibiting an open channel probability (n·Po) of 0.63, with open dwell time values τ0-1 τ0-2 and of 1.9 and 8.2 ms. After successive application of 3 μM tolbutamide (FIGS. 12B and 12E) and 30 μM tolbutamide (FIGS. 12C and 12F), n·Po decreased to 0.44 and 0.09, respectively, but the open dwell time values are not appreciably affected by the drug. Closed channel dwell times are increased in duration and frequency by tolbutamide (FIGS. 12H and 12I). Thus, the channel of the present inventions exhibits a form of channel inhibition in which the blocking compound had no effect on open channel dwell times and a progressive increase in long closures. This form of channel inhibition is similar to that produced by sulfonylureas acting on the KATP channel in pancreatic β cells. See, Gillis et. al., 1989; Babeenko et. al., 1999).
  • Example 12
  • Application of 100 μM of the SUR-activator diazoxide activates the 35pS channel of the present invention, causing weak inward rectification in cell-attached patches (FIGS. 13A, 13B and 13C). To determine the type of S U R affecting activation of the NCCa-ATP channel of the present invention, experiments are conducted using sulfonylurea compounds that preferentially activate SUR 2 over SUR1, namely cromakalin, and pinacidil. Both cromakalin and pinacidil had no effect on the NCCa-ATP channel of the present invention, which is consistent with other data described herein indicating that SUR1 is associated with the NCCa-ATP channel of the present invention, and activation of the channel is not mediated by SUR2.
  • Example 13 SUR-Mediated Cell Swelling
  • After addition of NaN3 to deplete ATP in cells, cell blebbing typically becomes apparent in 7-10 minutes. Diazoxide is an SUR1 agonist or SUR1 activator. When diazoxide alone is added to the cells, blebbing occurs even without ATP depletion, Diazoxide, therefore, opens the channel directly without ATP depletion by activating SUR1. However, when cells are pretreated with glibenclamide, addition of NaN3 does not cause blebbing, even after 30 minutes. Thus, activation of NCCa-ATP channel by ATP depletion or by the channel opener, diazoxide, can result in blebbing and swelling of NRAs, and that swelling can be prevented by blocking the channel with glibenclamide. ATP depletion by Na azide can result in necrotic cell death of NRAs. This finding accords with the data described herein that glibenclamide protects from the opening of the NCCa-ATP channel following ATP depletion, and that opening of this channel is responsible for cell blebbing.
  • The antagonist used in the methods of the present invention includes a compound that interferes with NCCa-ATP function. Typically, the effect of an antagonist is observed as a blocking of NCCa-ATP current in conditions under which the channel has been activated and current can be measured in the absence of the antagonist.
  • In addition to SUR1 specific sulfonylurea compounds, agents that block SUR1, also include compounds that are structurally unrelated to sulfonylureas. Such SUR1 blockers include a class of insulin secretagogues compounds that bind to the SUR, which were identified and developed for the treatment of type 2 diabetes. The benzamido derivatives: repaglinide, nateglinide, and meglitinide represent one such class of insulin secretagogues, that bind to the SUR. Nateglinide is an amino acid derivative. Also, imidazoline derivatives have been identified that interact with the sulfonylurea receptor (SUR) 1 subunit such as midaglizole (KAD-1229), LY397364 and LY389382.
  • In one preferred embodiment of the present invention, compounds that preferentially block SUR1, but not SUR2, are used in the method of the present invention. Such compounds include tolbutamide and gliclazide. The following compounds block both SUR1 and SUR2: glibenclamide, glimepiride, repaglinide, and meglitinide. In yet another embodiment of the method of the present invention, administration is combined with MgADP, which has been show to produce an apparent increase of sulfonylurea efficacy on channels containing SUR1, but not SUR2.
  • Example 14
  • To determine whether NCCa-ATP activation by ATP depletion initiates necrosis of reactive astrocytes that express this channel, studies are conducted to determine if glibenclamide is capable of protecting reactive astrocytes from cell death by inhibiting NCCa-ATP channel activity via its action on SUR1. Two types of cell death, apoptosis and necrosis, are assessed following ATP depletion.
  • Thus, activation of NCCa-ATP channel is responsible for necrotic death of NRAs following ATP depletion, and that glibenclamide can prevent this form of cell death.
  • In this Example, the preparation of freshly isolated NRAs was further purified by removal of RBCs, as described herein to provide a cell population having <1% RBCs. Over 95% of cells had resting potentials near EK, suggesting that the enzymatic dissociation method had not appreciably harmed the cells. Over 95% of cells are positive for the astrocyte marker, glial fibrillary acidic protein (GFAP) as determined by immunofluorescence. When examined by phase microscopy, the NRAs are of various sizes, ranging from 11-45 pms in diameter, some of which are phase bright and others are phase dark. A subgroup of phase bright cells had multiple short but distinct cell processes that are shorter than the cell soma. In this Example, only larger (≈30 μm diameter), phase bright cells with short processes (<1 cell length) are studied. This population of NRAs reliably express the NCCa-ATP channels.
  • Experiments are conducted at room temperature (22-25° C.) within 24 hr of cell isolation. An aliquot of cells is placed on a chamber slide (LAB-TEK, Naperville, Ill.) filled with extracellular bath solution containing (a): NaCl 130, KCl 10, CaCl 2 1, MgCl 2 1, HEPES 32.5, glucose 12.5, pH 7.4. After viable cells adhered to the surface, any residual debris not previously removed by centrifugation is washed away by flushing with excess solution. Cells are subjected to ATP depletion by 1 mM Na azide to activate (open) the NCCa-ATP channels, and then incubated with glibenclamide (1 μM).
  • Thereafter, the cells are examined by propidium iodide (PI) staining for evidence of cellular membrane permeabilization, an indication of early oncotic or necrotic cell death. See, Barros et al., 2001. The cells are also examined by fluorescein-tagged annexin V binding for evidence of externalization of the phosphoaminolipid phosphotidylserine from the inner face of the plasma membrane to the outer surface, an early indication of apoptosis. See, Clodi et al., 2000; Rucker-Martin et al., 1999. Staining procedure are conducting according to manufacture directions (Vybrant Apoptosis Assay Kit 2, Molecular Probes). Slides are mounted using ProLong antifade mounting medium (Molecular Probes). Signals are visualized using a Nikon Diaphot epifluorescent microscope (Leitz Wetzlar). Images are captured and stored using a SenSys digital camera (Roper Scientific Inc.) and IPLab software (version 3.0; Scanalytics Inc.). Annexin V-positive cells or PI-positive cells are counted in 20 individual fields using a 20× objective lens. Mean values of positive cells in 20 fields for various treatment groups are compared using ANOVA Pairwise multiple comparisons, with p<0.05 being considered as indicating a significant difference.
  • The fluorescence microscopy photos shown in FIG. 15A show that under baseline (control) conditions, both annexin V-positive and PI-positive cells (photos a and d, respectively) are rare in the cell isolates. After a 10-min incubation with Na aide (1 mM), the number of PI-positive cells increased substantially (p<0.05) (FIG. 15A at photo b and FIG. 15B). This indicates that ATP depletion triggers necrotic death in these cells. By contrast, Na azide treatment caused the number of annexin V-positive cells to increase slightly; the increase not being statically significant (p>0.05) (FIG. 15A at photo e and FIG. 15C). This indicates that apoptotic death was not a major endpoint of ATP depletion in these cells.
  • Pretreatment of cells with glibenclamide (1 μM) at the time of administration of Na aide dramatically decreased the number of PI-positive cells (p<0.05; FIG. 15A at photo c and FIG. 15B), indicating significant protection from necrotic death following ATP depletion. The number of NRAs undergoing apoptotic death also decreased with glibenclamide, as indicated by annexin V labeling (FIG. 15A at photo f and FIG. 15C), but values for this group were not significantly different.
  • This data indicate that the NCCa-ATP channel is involved in the mechanism of the necrotic cell death of reactive astrocytes. This Example shows that necrotic, rather than apoptotic, cell death is the principal endpoint of ATP depletion in these cells. Therefore, ATP depletion by Na azide initiates cell death by removal of the ATP block of the NCCa-ATP channel, thus initiating oncotic cell swelling. Involvement of this channel in oncotic cell swelling is confirmed by showing that necrotic death can also be induced by diazoxide, the channel opener that activates the NCCa-ATP channel in these cells, and could be blocked by glybenclamide, which prevents opening of the NCCa-ATP channel. The involvement of the NCCa-ATP channel in cell death of reactive astrocytes provides a mechanism and target of death in these cells, as well as the importance of blocking the NCCa-ATP channel to prevent the death of reactive astrocytes, which occurs in traumatic brain injury.
  • Example 15 In Vitro Assays for Determining a Test Compound Ability to Provide Dose-Dependent Blockage of the NCCa-ATP Channel
  • NCCa-ATP channels blocking compounds can be identified by a method in which the direct inhibitory effect of the test compound on NCCa-ATP channels is determined. Inside-out patches are used to show the inhibitory effect of the compound. To ensure that no K+ channel, particularly KATP is contributing to patch current, Cs+ is used as the charge carrier. Compounds that profoundly diminish channel activity, and the activity is shown to be due to a 35 pS cation channel, such a compound is identified as a compound that blocks the NCCa-ATP channels and is capable of inhibiting neuronal cell swelling and brain swelling. Varying concentrations of the compound are used to determine whether the NCCa-ATP channel is blocked by the compound in a dose-dependent manner. The concentration at which half maximum inhibition (EC50) is observed and the concentration at which channel activity is completely lost are determined. The sensitivity of the NCCa-ATP channel of the present invention to blocking in NRAs with the test compound can be compared. This in vitro assay for determining the concentration of the test compound which achieves a half-maximal inhibition of channel activity may be used to formulate dose in animal models to achieve a circulating plasma concentration range.
  • Example 16 In Vivo Assays for Determining Dose-Dependent Blockage of the NCCa-ATP Channel
  • The concentration of the test compound which achieves a half-maximal inhibition of channel activity is used to formulate dose in animal models to achieve a circulating plasma concentration range. The dose of test compound that achieves a circulating plasma concentration range calculated by methods known in the art is administered to an animal having brain injury or cerebral ischemia. To determine whether the test compound prevents, inhibits or diminishes brain swelling, the epidural pressure and/or intracranial pressure of the animal is measured, such as by using a microballoon, to quantitatively monitor brain swelling. Also, the swelling can be monitored by magnetic resonance (MR) imaging. Three different studies start administration prior to, at the time of, or after the brain injury. A compound that provided diminishes brain swelling, as compared to controls, is identified as a compound capable of inhibiting neuronal cell swelling and brain swelling. Varying concentrations of the compound are used to determine whether the compound delivers efficacy in a dose-dependent manner. The dose at which half maximum inhibition is observed and the concentration at which brain swelling is most quickly alleviated are determined. Formulations are produced comprising the optimal effective dose of the test compound for preventing, inhibiting, or diminishing brain swelling, along with a pharmaceutically acceptable carrier.
  • Summary of NCCa-ATP Channel Characteristics
  • The characteristics of cells expressing and composition containing the NCCa-ATP channel of the present invention are summarized in Table 1.
  • TABLE 1
    Properties of cells and
    membrane compositions
    containing the NCCa-ATP Membrane Preparation derived
    Channel of the Present from freshly isolated native reactive
    Invention Reactive Astrocytes astrocytes
    Monovalent cation permeable? Yes: Yes:
    NA+ NA+
    K+ K+
    Li+ Li+
    Rb+ Rb+
    Cs+ Cs+
    (NA+≈K+≈Li+≈Rb+) (NA+≈K+≈Li+≈Rb+)
    Anion permeable? No No
    Divalent cation permeable? No No
    Compounds blocking channel SUR1 antagonists SUR1 ANTAGONISTS
    activity
    Channel opening Intracell. ATP INTRACELL. ATP D
    Requires: depletion EPLETION
    Intracell. Mg2+ INTRACELL. MG2+
    Single Channel Conductance  ~35 pS  ~35 PS
    Activation <1.0 μM <1.0 MM
    [Ca2+]
    [ATP]1 EC50 (um) 0.79 μM 0.79 MM
    ADP No channel effect No channel effect
    AMP
    Pore radius 0.41 0.41
    (nm)
  • Methods of the Present Invention
  • The present invention provides a previously unknown ion channel found in mammalian neural cells that plays a role in cell swelling. The present invention further provides a method of screening for antagonists to the channel and a new use for antagonists to the channel, including sulfonylurea compounds such as glibenclamide and tolbutamide, as a treatment for brain swelling in mammals.
  • Methods of the present invention for identifying compounds that interact with, (e.g., bind to, open, block) the NCCa-ATP channel and employ (i) cell based assays and/or (ii) non-cell based assay systems. Such compounds may act as antagonists or agonists of NCCa-Ap channel activity. In a preferred embodiment of the present invention, antagonists that block and/or inhibit the permeability of the NCCa-ATP channel are utilized in methods for treating neural cell swelling and/or brain swelling.
  • The cell based assays use neural cells that express the NCCa-ATP channel, preferably a functional NCCa-ATP channel; the preferred cells are “MS. The non-cell based assay systems include membrane preparations that express the NCCa-ATP channel, preferably a functional NCCa-ATP channel. Cell-based assays include, but are not limited to, compound binding assays, microscopic observation of cell status (normal, blebbing, swelling), and measuring channel currents both before and after exposure to compound. Compositions comprising membrane preparations expressing the NCCa-ATP channel may be used to identify compounds that interact with, bind to, block or open the NCCa-ATP channel or SUR1. The term “expressing the NCCa-ATP channel” or “expresses the NCCa-ATP channel” means having a functional NCCa-ATP channel. The term “functional NCCa-ATP channel” as used herein means an NCCa-ATP channel capable of being detected. One preferred method of detecting the NCCa-ATP channel is by determining, in vitro or in vivo, whether the channel is open, closed and/or blocked.
  • For example, in a typical experiment using a membrane preparation, NRAs that express the NCCa-ATP channel are used to produce the membrane preparation. Methods for producing membranes from whole cells and tissues are well known in the art. One such method produces purified cell membranes in the form of a purified microsomal fraction isolated from disrupted cells or a tissue sample by discontinuous sucrose gradient centrifugation. Also included are membranes comprised of cell-attached patches, inside-out patches, or outside-out patches. One example of a tissue sample expressing NCCa-ATP channels is brain tissue adjacent to brain injury.
  • The membranes preparations are used in a number of assays, including, but not limited to measuring channel currents, both before and after exposure to compound; and in vitro binding assays. The experimental conditions for such assays to determine and quantify the status of the NCCa-ATP channel are described throughout the instant specification, including binding assay conditions, bath compositions, pipette solutions, concentrations of ATP and Ca2+ required, membrane voltage, membrane potentials, compound quantity ranges, controls, etc.
  • Binding assays and competitive binding assays employ a labeled ligand or antagonist of the NCCa-ATP channel. In one such experiment, labeled Glibenclamide, such as FITC-conjugated glibenclamide or radioactively labeled glibenclamide is bound to the membranes and assayed for specific activity; specific binding is determined by comparison with binding assays performed in the presence of excess unlabelled antagonist.
  • In one method for identifying NCCa-ATP channel blockers, membranes are incubated with a labeled compound shown to block this channel, in either the presence or absence of test compound. Compounds that block the NCCa-ATP channel and compete with the labeled compound for binding to the membranes will have a reduced the signal, as compared to the vehicle control samples. In another aspect of the invention the screens may be designed to identify compounds that compete with the interaction between NCCa-ATP channel and a known (previously identified herein) NCCa-ATP channel antagonist or SUR1 antagonist, such as glibenclamide. In such screens, the known NCCa-ATP channel antagonist or SUR1 antagonist is labeled and the test compounds are then assayed for their ability to compete with or antagonize the binding of the labeled antagonist.
  • The assays described herein can be used to identify compounds that modulate or affect NCCa-ATP channel activity. For example, compounds that affect NCCa-ATP channel activity include but are not limited to compounds that bind to the NCCa-ATP channel or SUR1, inhibit binding of identified blockers or ligands (such as glibenclamide), and either open/activate the channel (agonists) or block/inhibit the channel (antagonists).
  • Assays described can also identify compounds that modulate neural cell swelling (e.g., compounds which affect other events involved in neural cell swelling that are activated by ligand binding to or blocking of the NCCa-ATP channel).
  • Compounds Screened in Accordance with the Invention
  • The compounds for screening in accordance with the invention include, but are not limited to organic compounds, peptides, antibodies and fragments thereof, peptidomimetics, that bind to the NCCa-ATP channel and either open the channel (i.e., agonists) or block the channel (i.e., antagonists). For use in the treatment of neural cell swelling or brain swelling, compounds that block the channel are preferred. Agonists that open or maintain the channel in the open state include peptides, antibodies or fragments thereof, arid other organic compounds that include the SUR1 subunit of the NCCa-ATP channel (or a portion thereof) and bind to and “neutralize” circulating ligand for SUR1.
  • With reference to screening of compounds that affect the NCCa-ATP channel, libraries of known compounds can be screened, including natural products or synthetic chemicals, and biologically active materials, including proteins, for compounds which are inhibitors or activators. Preferably, such a compound is an NCCa-ATP antagonist, which includes an NCCa-ATP channel inhibitor, an NCCa-ATP channel blocker, a SUR1 antagonist, SUR1 inhibitor, and/or a compound capable of reducing the magnitude of membrane current through the channel.
  • Compounds may include, but are not limited to, small organic or inorganic molecules, compounds available in compound libraries, peptides such as, for example, soluble peptides, including but not limited to members of random peptide libraries; (see, e.g., Lam, K. S. et al., 1991, Nature 354: 82-84; Houghten, R. et al., 1991, Nature 354: 84-86), and combinatorial chemistry-derived molecular library made of D- and/or L-configuration amino acids, phosphopeptides (including, but not limited to, members of random or partially degenerate, directed phosphopeptide libraries; see, e.g., Songyang, Z. et al., 1993, Cell 72: 767-778), antibodies (including, but not limited to, polyclonal, monoclonal, humanized, anti-idiotypic, chimeric or single chain antibodies, and FAb, F(ab′).sub.2 and FAb expression library fragments, and epitope-binding fragments thereof).
  • Other compounds which can be screened in accordance with the invention include but are not limited to small organic molecules that are able to cross the blood-brain barrier, gain entry into an appropriate neural cell and affect the expression of the NCCa-ATP channel gene or some other gene involved in the NCCa-ATP channel activity (e.g., by interacting with the regulatory region or transcription factors involved in gene expression); or such compounds that affect the activity of the NCCa-ATP channel or the activity of some other intracellular factor involved in the NCCa-ATP channel activity.
  • Computer modeling and searching technologies permit identification of compounds, or the improvement of already identified compounds, that can modulate NCCa-ATP channel activity or expression. Having identified such a compound or composition, the active sites or regions are identified. Such active sites might typically be ligand binding sites. The active site can be identified using methods known in the art including, for example, from study of complexes of the relevant compound or composition with other ligands, from the amino acid sequences of peptides, or from the nucleotide sequences of nucleic acids. Chemical or X-ray crystallographic methods can be used to study complexes of the relevant compound to find the active site. The three dimensional geometric structure of the active site is determined. This can be done by known methods, including X-ray crystallography, which can determine a complete molecular structure. On the other hand, solid or liquid phase NMR can be used to determine certain intra-molecular distances. Any other experimental method of structure determination can be used to obtain partial or complete geometric structures. The geometric structures may be measured with a complexed ligand, natural which may increase the accuracy of the active site structure determined.
  • If an incomplete or insufficiently accurate structure is determined, the methods of computer based numerical modeling can be used to complete the structure or improve its accuracy. Any recognized modeling method may be used, including parameterized models specific to particular biopolymers such as proteins or nucleic acids, molecular dynamics models based on computing molecular motions, statistical mechanics models based on thermal ensembles, or combined models. For most types of models, standard molecular force fields, representing the forces between constituent atoms and groups, are necessary, and can be selected from force fields known in physical chemistry. The incomplete or less accurate experimental structures can serve as constraints on the complete and more accurate structures computed by these modeling methods.
  • Finally, having determined the structure of the active site, either experimentally, by modeling, or by a combination, candidate modulating compounds can be identified by searching databases containing compounds along with information on their molecular structure. Such a search seeks compounds having structures that match the determined active site structure and that interact with the groups defining the active site. Such a search can be manual, but is preferably computer assisted. These compounds found from this search are potential NCCa-ATP channel modulating, preferably blocking, compounds.
  • Alternatively, these methods can be used to identify improved modulating compounds from an already known modulating compound or ligand. The composition of the known compound can be modified and the structural effects of modification can be determined using the experimental and computer modeling methods described above applied to the new composition. The altered structure is then compared to the active site structure of the compound to determine if an improved fit or interaction results. In this manner systematic variations in composition, such as by varying side groups, can be quickly evaluated to obtain modified modulating compounds or ligands of improved specificity or activity.
  • Examples of molecular modeling systems are the CHARMm and QUANTA programs (Polygen Corporation, Waltham, Mass.). CHARMm performs the energy minimization and molecular dynamics functions. QUANTA performs the construction, graphic modeling and analysis of molecular structure. QUANTA allows interactive construction, modification, visualization, and analysis of the behavior of molecules with each other. A number of articles review computer modeling of drugs interactive with specific proteins, such as Rotivinen, et al.) 1988, Acta Pharmaceutical Fennica 97: 159-166); Ripka (1988 New Scientist 54-57); McKinaly and Rossmann (1 989, Annu. Rev. Pharmacol. Toxicol. 29: 11 1-122); Perry and Davies, OSAR: Quantitative Structure-Activity Relationships in Drug Design pp. 189-193 Alan R. Liss, Inc. 1989; Lewis and Dean (1989, Proc. R. SOC. Lond. 236: 125-140 and 141-162); and, with respect to a model receptor for nucleic acid components, Askew, et al. (1989, J. Am. Chem. SOC. 11 1: 1082-1 090). Other computer programs that screen and graphically depict chemicals are available from companies such as BioDesign, Inc. (Pasadena, Calif.), Allelix, Inc. (Mississauga, Ontario, Canada), and Hypercube, Inc. (Cambridge, Ontario). Although these are primarily designed for application to drugs specific to particular proteins, they can be adapted to design of drugs specific to regions of DNA or RNA, once that region is identified.
  • Compounds identified via assays such as those described herein may be useful, for example, in elaborating the biological function of the NCCa-ATP channel and for relief of brain swelling.
  • Assays for testing the efficacy of compounds identified in the cellular screen can be tested in animal model systems for brain swelling. Such animal models may be used as test substrates for the identification of drugs, pharmaceuticals, therapies and interventions which may be effective in treating brain swelling. For example, animal models of brain swelling, such as brain injury, may be exposed to a compound, suspected of exhibiting an ability to inhibit brain swelling, at a sufficient concentration and for a time sufficient to elicit such an inhibition of brain swelling in the exposed animals. The response of the animals to the exposure may be monitored using visual means (e.g., radiological, CAT, MRI), measurement of intracranial pressure, and/or the reversal of symptoms associated with brain swelling. With regard to intervention, any treatments which reverse any aspect of brain swelling-associated symptoms should be considered as candidates for brain swelling therapeutic intervention. Dosages of test agents may be determined by deriving dose-response curves, as discussed herein.
  • Accordingly, the present invention is useful in the treatment or alleviation of neural cell swelling and death and brain swelling, especially those brain insults related to traumatic brain injury, central or peripheral nervous system damage, cerebral ischemia, such as stroke, or complications involving and/or stemming from edema, injury, or trauma. Such damage or complications may be characterized by an apparent brain damage or aberration, the symptoms of which can be reduced by the methods of the present invention including the administration of an effective amount of the active compounds or substances described herein. According to a specific embodiment of the present invention the administration of effective amounts of the active compound can block the channel, which if remained open leads to neural cell swelling and cell death. A variety of antagonists to SUR1 are suitable for blocking the channel. Examples of suitable SUR1 antagonists include, but are not limited to glibenclamide, tolbutamide, repaglinide, nateglinide, meglitinide, midaglizole, LY397364, LY3 89382, gliclazide, glimepiride, MgADP, and combinations thereof. In a preferred embodiment of the invention the SUR1 antagonists is selected from the group consisting of glibenclamide and tolbutamide. Still other therapeutic “strategies” for preventing neural cell swelling and cell death can be adopted including, but not limited to methods that maintain the neural cell in a polarized state and methods that prevent strong depolarization.
  • Example 17 Additional Mechanisms for Maintaining NRAS in a Polarized State
  • When reactive astrocytes are strongly depolarized due to opening of the NCCa-ATP channel, they undergo blebbing and swelling and eventually sustain necrotic cell death. As stated above, when reactive astrocytes are strongly depolarized due to opening of a non-selective channel that is sensitive to Ca2+ and ATP (NCCa-ATP channel), they undergo blebbing and swelling and eventually sustain necrotic cell death. The death of these reactive astrocytes can be prevented if strong depolarization can be prevented, in other words, if the cells can be maintained in a polarized state.
  • One potential way of maintaining the NRAs in a polarized state is to open the Kir2.3 channel. NRAs are exposed to the Kir2.3 channel opener, Tenidap, to maintain Kir2.3 channels open. Native reactive astrocytes freshly harvested from adult rat brains after injury are exposed to Tenidap to evaluate the drug's ability to open the Kir2.3 channel in these cells. Preferably, type 1 reactive (R1) astrocytes are harvested and used in this assay. One of the subtypes of reactive astrocytes is the type R1 astmcyte. Type R1 astrocytes comprise the largest population of recoverable astrocytes at the site of brain injury. They are characteristically located in the region of tissue surrounding the injury site, many of which are found to have migrated into the injury site itself. See, Perillan, et al., 1999.
  • The reactive astrocytes that are part of the cellular response to TBI and stroke are comprised of at least two subtypes. One of the subtypes of reactive astrocytes is the type R1 astrocyte. Type R1 astrocytes comprise the largest population of recoverable astrocytes at the site of brain injury. They are characteristically located in the region of tissue surrounding the injury site, with many of these cells also being found to have migrated into the injury site itself. See, Perillan, et al. 1999.
  • Type R1 astrocytes are the predominant type of reactive astrocyte in the NRA preparations. Type R1 astrocytes express two critically important ion channels in their cell membrane: (a) the Kir2.3 channel, which is present in cultured as well as freshly isolated cells; and (b) the NCCa-ATP channel, which is present only in freshly isolated reactive astrocytes and lost shortly after culturing. The Kir2.3 is an inward rectifier channel that is critically important for maintaining the cell polarized to a normal resting potential near the potassium reversal potential (≈−75 mV). When this channel is inactivated or inhibited, the cell depolarizes to a potential near the chloride reversal potential (≈−25 mV). Characteristic features of the NCCa-ATP channel are: 1) it is a non-selective cation channels that allows passage of Nay K and other monovalent cations quite readily; 2) it is activated by an increase in intracellular calcium, and/or by a decrease in intracellular ATP; and 3) it is regulated by sulfonylurea receptor type 1 (SUR1). SUR1 had been considered to be associated exclusively with KATP channels, such as those found in pancreatic cells.
  • Opening of the NCCa-ATP channel following ATP depletion, as with ischemia or hypoxia, causes depolarization of the cell due to influx of Na. This influx of Na increases the osmotic load within the cell, and as a result, H 20 enters the cell to equilibrate the osmotic load. The result is an excess of Na and H 20 intracellularly, a pathological response that produces cell blebbing and cell swelling and that is known as cytotoxic edema. Left unchecked, this pathological response eventually leads to cell death. As disclosed herein, this cell death is mostly necrotic cell death but to a lesser extent, apoptotic cell death as well.
  • A number of approaches may be used to meliorate brain swelling due to cytotoxic edema. One currently used treatment for treating patients in relevant clinical situations is based on increasing extracellular osmolarity to reduce the driving force for influx of H 20. This strategy also reduces blebbing in isolated cells.
  • A more specific strategy to reduce cytotoxic edema is inactivating or blocking the NCCa-ATP channel that is primarily responsible for the influx of Na that draws H 20 into the cell and that actually causes cytotoxic edema. One highly selective approach to inactivating this channel is to exploit the unique relationship between the channel and the controlling regulatory subunit, SUR1. A variety of drugs have been developed that interact with SUR1 in pancreatic p cells to block the KATP channel in those cells and thereby treat diabetes. Some of these drugs belong to the class of agents called sulfonylureas. As described herein, drugs that block the KATP channel, such as glybenclamide and tolbutamide, are highly effective at blocking the NCCa-ATP channel in type R1 astrocytes. Drugs capable NCCa-ATP channel blocking in NRAs (a) prevents cell blebbing in response to ATP depletion, (b) significantly reduces cell death following ATP depletion. Also, the use of glybenclamide to treat brain swelling in an animal suffering from stroke or brain injury is described herein.
  • Yet another strategy to oppose the effect of the NCCa-ATP channel and reduce cytotoxic edema would be to counteract depolarization of the cell that accompanies opening of the NCCa-ATP channel. One way to accomplish this is to enhance opening of the Kir2.3 channels that are also present in these cells. An anti-inflammatory compound, Tenidap, is an opener of Kir2.3 channels. See, Popp et al., 1992; Liu et al., 2002. [OO1SO] Tenidap is evaluated for its ability to reduce cell blebbing and swelling and necrotic cell death in response to ATP depletion in the isolated cells as well as in situ in injured rat brain. To assess whether Tenidap opens the Kir2.3 channels in type R1 astrocytes, using methods similar to those described herein for evaluating the status of the NCCa-ATP channel. Results from such experiments that show Tenidap to open Kir2.3 channels in type R1 astrocytes, and reduce cell blebbing and cell death in response to ATP depletion would indicate the usefulness of Tenidap in treating brain swelling and cytotoxic edema resulting from TBI or cerebral ischemia. The effective amount of Tenidap is that amount capable of reducing brain swelling or cerebral ischemia due to the drug's ability to inhibit neural cell swelling and necrotic cell death.
  • SUR1 blockers are likely to be the most specific, reliable provide the fewest untoward side effects. Further, a combination of treatments including use of osmotic diuretics, NCCa-ATP channel blockers such glybenclamide and Kir2.3 channel openers such as Tenidap may provide better efficacy in ameliorating cytotoxic edema and reducing morbidity and mortality in brain injury and stroke.
  • It is therefore another object of the present invention to provide a method for identifying compounds that inhibit neural cell swelling, comprising: (a) contacting a test compound with a composition comprising the Kir2.3 channel, and (b) determining whether the test compound opens the Kir2.3 channel, wherein a test compound that opens the Kir2.3 channel is identified as a compound for inhibiting neural cell swelling.
  • It is yet another object of the present invention to provide a method for a method for identifying compounds that inhibit brain swelling, comprising: (a) contacting a test compound with a composition comprising the Kir2.3 channel, and (b) determining whether the test compound opens the Kir2.3 channel, wherein a test compound that opens the Kir2.3 channel is identified as a compound for inhibiting brain swelling.
  • It is yet another object of the present invention to provide a method for a method for identifying compounds that inhibit neural cell swelling and/or brain swelling in an animal, comprising: (a) contacting a test compound with a composition comprising the Kir2.3 channel, and (b) determining whether the test compound opens the Kir2.3 channel, wherein a test compound that opens the Kir2.3 channel is identified as a compound for inhibiting neural cell swelling and/or brain swelling in an animal.
  • It is a further object of the present invention to provide a method for identifying compounds that prevent, inhibit and/or alleviate brain swelling in a subject, comprising: (a) contacting a test compound with a composition comprising the Kir2.3 channel, and determining whether the test compound opens the Kir2.3 channel, and (b) administering the test compound to a subject having a brain injury or cerebral ischemia, and determining whether the test compound prevents, inhibits and/or alleviates brain swelling in the subject, wherein test compounds that open the Kir2.3 channel are identified as compounds that inhibit brain swelling.
  • It is a further object of the present invention to provide a method for identifying compounds that inhibit neural cell swelling in an animal, comprising: (a) contacting a test compound with a composition comprising the Kir2.3 channel, and determining whether the test compound opens the Kir2.3 channel, and (b) administering the test compound to an animal having a brain injury or cerebral ischemia, and determining whether the test compound inhibits brain swelling of the treated animal, wherein test compounds that inhibit brain swelling are identified as compounds that inhibit neural cell swelling in an animal.
  • It is also an object of the present invention to provide a method of preventing neural cell swelling in the brain of a subject, said method comprising administering to the subject a formulation containing an effective amount of a compound that opens the Kir2.3 channel and a pharmaceutically acceptable carrier.
  • It is a further objection of the present invention to provide a method of alleviating the negative effects of traumatic brain injury or cerebral ischemia stemming from neural cell swelling in a subject, comprising administering to the subject a formulation comprising an effective amount of a compound that opens the Kir2.3 channel and a pharmaceutically acceptable carrier. In the object of the present invention that provide methods assessing the effect of a compound on the Kir2.3 channel, a preferred compound is Tenidap. For example the formulation may provide a daily dose of Tenidap that is from about 10 mg/day to about 500 mg/day, or, when administered directly to the brain the daily dose of Tenidap is from about 500 mg/day to 1.5 gms/day or greater.
  • Pharmaceutical Formulations and Methods of Treating Neural Cell Swelling and Brain Swelling Compositions of the Present Invention
  • The present invention also contemplates therapeutic methods employing compositions comprising the active substances disclosed herein. Preferably, these compositions include pharmaceutical compositions comprising a therapeutically effective amount of one or more of the active compounds or substances along with a pharmaceutically acceptable carrier.
  • As used herein, the term “pharmaceutically acceptable” carrier means a non-toxic, inert solid, semi-solid liquid filler, diluent, encapsulating material, formulation auxiliary of any type, or simply a sterile aqueous medium, such as saline. Some examples of the materials that can serve as pharmaceutically acceptable carriers are sugars, such as lactose, glucose and sucrose, starches such as corn starch and potato starch, cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt, gelatin, talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol, polyols such as glycerin, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate, agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline, Ringer's solution; ethyl alcohol and phosphate buffer solutions, as well as other non-toxic compatible substances used in pharmaceutical formulations.
  • Wetting agents, emulsifiers and lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator. Examples of pharmaceutically acceptable antioxidants include, but are not limited to, water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfite, sodium metabisulfite, sodium sulfite, and the like; oil soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, aloha-tocopherol and the like; and the metal chelating agents such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid and the like.
  • Dose Determinations
  • By a “therapeutically effective amount” or simply “effective amount” of an active compound, such as glibenclamide or tolbutamide, is meant a sufficient amount of the compound to treat or alleviate the brain swelling at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the active compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the brain injury or ischemia; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coinciding with the specific compound employed; and like factors well known in the medical arts.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell assays or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell based assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
  • The total daily dose of the active compounds of the present invention administered to a subject in single or in divided doses can be in amounts, for example, from 0.01 to 25 mg/kg body weight or more usually from 0.1 to 15 mg/kg body weight. Single dose compositions may contain such amounts or submultiples thereof to make up the daily dose. In general, treatment regimens according to the present invention comprise administration to a human or other mammal in need of such treatment from about 1 mg to about 1000 mg of the active substance(s) of this invention per day in multiple doses or in a single dose of from 1 mg, 5 mg, 10 mg, 100 mg, 500 mg or 1000 mg.
  • In certain situations, it may be important to maintain a fairly high dose of the active agent in the blood stream of the patient, particularly early in the treatment. Such a fairly high dose may include a dose that is several times greater than its use in other indications. For example, the typical anti-diabetic dose of oral or IV glibenclamide is about 2.5 mg/kg to about 15 mg/kg per day; the typical anti-diabetic dose of oral or IV tolbutamide is about to 0.5 gm/kg to about 2.0 gm/kg per day; the typical anti-diabetic dose for oral gliclazide is about 30 mg/kg to about 120 mg/kg per day; however, much larger doses may be required to block neural cell swelling and brain swelling.
  • For example, in one embodiment of the present invention directed to a method of preventing neural cell swelling in the brain of a subject by administering to the subject a formulation containing an effective amount of a compound that blocks the NCCa-ATP channel and a pharmaceutically acceptable carrier; such formulations may contain from about 0.1 to about 100 grams of tolbutamide or from about 0.5 to about 150 milligrams of glibenclamide. In another embodiment of the present invention directed to a method of alleviating the negative effects of traumatic brain injury or cerebral ischemia stemming from neural cell swelling in a subject by administering to the subject a formulation containing an effective amount of a compound that blocks the NCCa-ATP channel and a pharmaceutically acceptable carrier.
  • In situations of traumatic brain injury or cerebral ischemia (such as stroke), or cerebral hypoxia, it may be important to maintain a fairly high dose of the active agent to ensure delivery to the brain of the patient, particularly early in the treatment. Hence, at least initially, it may be important to keep the dose relatively high and/or at a substantially constant level for a given period of time, preferably, at least about six or more hours, more preferably, at least about twelve or more hours and, most preferably, at least about twenty-four or more hours. In situations of traumatic brain injury or cerebral ischemia (such as stroke), it may be important to maintain a fairly high dose of the active agent to ensure delivery to the brain of the patient, particularly early in the treatment.
  • When the method of the present invention is employed to treat conditions involving bleeding in the brain, such as traumatic brain injury or cerebral ischemia (such as stroke), delivery via the vascular system is available and the compound is not necessarily required to readily cross the blood-brain barrier.
  • Formulations and Administration
  • The compounds of the present invention may be administered alone or in combination or in concurrent therapy with other agents which affect the central or peripheral nervous system, particularly selected areas of the brain.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs containing inert diluents commonly used in the art, such as water, isotonic solutions, or saline. Such compositions may also comprise adjuvants, such as wetting agents; emulsifying and suspending agents; sweetening, flavoring and perfuming agents.
  • Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
  • The injectable formulation can be sterilized, for example, by filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions, which can be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.
  • In order to prolong the effect of a drug, it is often desirable to slow the absorption of a drug from subcutaneous or intramuscular injection. The most common way to accomplish this is to inject a suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug becomes dependent on the rate of dissolution of the drug, which is, in turn, dependent on the physical state of the drug, for example, the crystal size and the crystalline form. Another approach to delaying absorption of a drug is to administer the drug as a solution or suspension in oil. Injectable depot forms can also be made by forming microcapsule matrices of drugs and biodegradable polymers, such as polylactide-polyglycoside. Depending on the ratio of drug to polymer and the composition of the polymer, the rate of drug release can be controlled. Examples of other biodegradable polymers include polyorthoesters and polyanhydrides. The depot injectables can also be made by entrapping the drug in liposomes or microemulsions, which are compatible with body tissues.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable non-irritating excipient, such as cocoa butter and polyethylene glycol which are solid at ordinary temperature but liquid at the rectal temperature and will, therefore, melt in the rectum and release the drug.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, gelcaps and granules. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such as magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings and other release-controlling coatings.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferably, in a certain part of the intestinal tract, optionally in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention further include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. Transdermal patches have the added advantage of providing controlled delivery of active compound to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel. The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • The method of the present invention employs the compounds identified herein for both in vitro and in vivo applications. For in vivo applications, the invention compounds can be incorporated into a pharmaceutically acceptable formulation for administration. Those of skill in the art can readily determine suitable dosage levels when the invention compounds are so used.
  • As employed herein, the phrase “suitable dosage levels” refers to levels of compound sufficient to provide circulating concentrations high enough to effectively block the NCCa-ATP channel and prevent or reduce neural cell swelling in vivo.
  • In accordance with a particular embodiment of the present invention, compositions comprising at least one SUR1 antagonist compound (as described above), and a pharmaceutically acceptable carrier are contemplated.
  • Exemplary pharmaceutically acceptable carriers include carriers suitable for oral, intravenous, subcutaneous, intramuscular, intracutaneous, and the like administration. Administration in the form of creams, lotions, tablets, dispersible powders, granules, syrups, elixirs, sterile aqueous or non-aqueous solutions, suspensions or emulsions, and the like, is contemplated.
  • For the preparation of oral liquids, suitable carriers include emulsions, solutions, suspensions, syrups, and the like, optionally containing additives such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents, and the like.
  • For the preparation of fluids for parenteral administration, suitable carriers include sterile aqueous or non-aqueous solutions, suspensions, or emulsions. Examples of non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate. Such dosage forms may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. They may be sterilized, for example, by filtration through a bacteria-retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They can also be manufactured in the form of sterile water, or some other sterile injectable medium immediately before use. The active compound is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • The foregoing disclosure of the preferred embodiments of the present invention has been presented for purposes of illustration and description. It is not intended to be exhaustive or to limit the invention to the precise forms disclosed. Many variations and modifications of the embodiments described herein will be apparent to one of ordinary skill in the art in light of the above disclosure. The scope of the invention is to be defined only by the claims appended hereto, and by their equivalents.
  • Further, in describing representative embodiments of the present invention, the specification may have presented the method and/or process of the present invention as a particular sequence of steps. However, to the extent that the method or process does not rely on the particular order of steps set forth herein, the method or process should not be limited to the particular sequence of steps described. As one of ordinary skill in the art would appreciate, other sequences of steps may be possible. Therefore, the particular order of the steps set forth in the specification should not be construed as limitations on the claims. In addition, the claims directed to the method and/or process of the present invention should not be limited to the performance of their steps in the order written, and one skilled in the art can readily appreciate that the sequences may be varied and still remain within the spirit and scope of the present invention. Further, in describing representative embodiments of the present invention, the specification may have presented the method and/or process of the present invention as a particular sequence of steps. However, to the extent that the method or process does not rely on the particular order of steps set forth herein, the method or process should not be limited to the particular sequence of steps described. As one of ordinary skill in the art would appreciate, other sequences of steps may be possible. Therefore, the particular order of the steps set forth in the specification should not be construed as limitations on the claims. In addition, the claims directed to the method and/or process of the present invention should not be limited to the performance of their steps in the order written, and one skilled in the art can readily appreciate that the sequences may be varied and still remain within the spirit and scope of the present invention.
  • All references cited herein are incorporated by reference in their entirety herein. Full citations for the references cited herein are provided in the following list.
    • Adams D J, Dwyer T M, Hille B (1980) The permeability of endplate channels to monovalent and divalent metal cations. J Gen Physiol 75: 493-510.
    • Aguilar-Bryan L., Nichols C G, Wechsler S W, Clement J P, Boyd A E, 111.
    • Gonzalez G, Herrera-Sosa H, Nguy K, Bryan J, Nelson D A (1995) Cloning of the beta cell high-affinity sulfonylurea receptor: a regulator of insulin secretion. Science 268: 423-426.
    • Barros L F, Stutzin A, Calixto A, Catalan M, Castro J, Hetz C, Hermosilla T (2001) Nonselective cation channels as effectors of free radical-induced rat liver cell necrosis. Hepatology 33: 114-122.
    • Bordey A, Sontheimer H (1998) Properties of human glial cells associated with epileptic seizure foci. Epilepsy Res 32: 286-303.
    • Brismar T, Collins V P (1993) Effect of external cation concentration and metabolic inhibitors on membrane potential of human glial cells. J Physiol (Lond) 460: 365-383.
    • Champigny G, Verrier B, Lazdunski M (1991) A voltage, calcium, and ATP sensitive non selective cation channel in human colonic tumor cells. Biochem Biophys Res Cornmun 176: 1 196-1 203.
    • Christensen O, Hofbann E K (1992) Cell swelling activates K+ and Cl− channels as well as nonselective, stretch-activated cation channels in Ehrlich ascites tumor cells. J Membr Biol 129: 13-36.
    • Chuang H, Jan Y N, Jan L Y (1997) Regulation of IRK3 inward rectifier K+ channel by ml acetylcholine receptor and intracellular magnesium. Cell 89: 1 121-1 132.
    • Cook D I, Poronnik P, Young J A (1990) Characterization of a 25-pS nonselective cation channel in a cultured secretory epithelial cell line. J Membr Biol 114: 37-52.
    • Fujita A, Kurachi Y (2000) Molecular aspects of ATP-sensitive K+ channels in the cardiovascular system and K+ channel openers. Pharmacol Ther 2000 January: 85 (1):39-53.
    • Gray M A, Argent B E (1990) Non-selective cation channel on pancreatic duct cells, Biochim Biophys Acta 1029: 33-42.
    • Hamill O P, Marty A, Neher E, Salunann B, Sigworth F J (1981) Improved patch-clamp techniques for high-resolution current recording from cells and cell-free membrane patches. Pflugers Arch 391: 85-100.
    • Harvey J, Hardy S C, Ashford M L (1999) Dual actions of the metabolic inhibitor, sodium azide on K(ATP) channel currents in the rat CRI-G1 insulinoma cell line. Br J Pharmacol 126: 51-60.
    • Higgins C F (1992) ABC transporters: from microorganisms to man. Annu Rev Cell Biol 8: 67-1 13.
    • Horn R, Marty A (1988) Muscarinic activation of ionic currents measured by a new whole-cell recording method. J Gen Physiol 92:145-159.
    • Inagaki N, Gonoi T, Clement J P, Wang C Z, Aguilar-Bryan L, Bryan J, Seino S (1996) A family of sulfonylurea receptors determines the pharmacological properties of ATP-sensitive K+ channels. Neuron 16:1011-1017.
    • Isomoto S, Kondo C, Yamada M, Matsumoto S, Higashiguchi O, Horio Y, Matsuzawa Y, Kurachi Y (1996) A novel sulfonylurea receptor forms with BIT (Kir6.2) a smooth muscle type ATP-sensitive K+ channel. J Biol Chem 271: 24321-24324.
    • Johnson M E, Gores G J, Uhl C B, Sill J C (1994) Cytosolic free calcium and cell death during metabolic inhibition in a neuronal cell line. J Neurosci 14: 4040-4049.
    • Jurkowitz-Alexander M S, Altschuld R A, Ham S E, Stephens R E, Horrocks L A (1993) Protection of ROC-1 hybridglial cells by polyethylene glycol following ATP depletion. J Neurochem 61:1581-1584.
    • Jurkowitz-Alexander M S, Altschuld R A, Hohl C M, Johnson J D, McDonald J S, Simmons T D, Horrocks L A (1992) Cell swelling, blebbing, and death are dependent on ATP depletion and independent of calcium during chemical hypoxia in a glial cell line (ROC-1). J Neurochem 59: 344-352.
    • Juurlink B H, Chen Y, Hertz L (1992) Can J Physiol Pharmacol 70 Suppl: S344-S349. Kempski O, von Rosen S, Weigt H, Staub F, Peters J, Baethmann A (1991). Ann N Y Acad Sci 633: 306-317.
    • Kim D, Fu C (1993) Activation of a nonselective cation channel by swelling in atrial cells. J Membr Biol 135: 27-37.
    • Kimelberg H K, Rutledge E, Goderie S, Charniga C (1995) Astrocytic swelling due to hypotonic or high K+ medium causes inhibition of glutamate and aspartate uptake and increases their release. J Cereb Blood Flow Metab 15: 409-4 16.
    • Kimelberg H K, Rose J W, Barron K D, Waniewski R A, Cragoe E J. (1989) Astrocytic swelling in traumatic-hypoxic brain injury. Beneficial effects of an inhibitor of anion exchange transport and glutamate uptake in glial cells. Mol Chem Neuropathol 11(1): 1-31.
    • Korbmacher C, Volk T, Segal A S, Boulpaep E L, Fromter E (1995) A calcium-activated and nucleotide-sensitive nonselective cation channel in M−1 mouse cortical collecting duct cells. Membr Biol 146: 29-45.
    • Kom S J, Marty A, Comer J A, Horn R (1991) Perforated patch recording. In: Methods in Neuroscience. Electrophysiology and Microinjection. (Conn P M, ed), pp 364-373. San Diego: Academic Press.
    • Lawson K (2000) Potassium channel openers as potential therapeutic weapons in ion channel disease. Kidney Int 2000 March: 57 (3): 838-845.
    • Lebovitz H E (1985) Oral hypoglycaemic agents. Amsterdam: Elsevier.
    • Liu Y, Liu D, Printzenhoff D, Coghlan M J, Harris R, Krafte D S (2002) Tenidap, a novel anti-inflammatory agent, is an opener of the inwardly rectifying K+ channel hKir2.3. Eur. J. Pharmacol. 435: 153-160.
    • Lomneth R, Gruenstein E I (1989) Energy-dependent cell volume maintenance in UC-11MG human astrocytornas. Am J Physiol 257: C817-C824.
    • Majno G, Joris I (1995) Apoptosis, oncosis, and necrosis. An overview of cell death. Am Path 01 146: 3-15.
    • Maruyama Y, Petersen O H (1984) Single calcium-dependent cation channels in mouse pancreatic acinar cells. J Membr Biol 81: 83-87.
    • Mongin A A, Cai 2, Kimelberg H K (1999) Volume-dependent taurine release from cultured astrocytes requires permissive [Ca2+]i and calmodulin. Am J Physiol 277: C823-C832.
    • Nichols C G, Shyng S L, Nestorowicz A, Glaser B, Clement J P, Gonzalez G, Aguilar-Bryan L, Permutt M A, Bryan J (1996) Adenosine diphosphate as an intracellular regulator of insulin secretion. Science 272: 1785-1787.
    • Ono S, Mougouris T, DuBose T D, Jr., Sansom S C (1994) ATP and calcium modulation of nonselective cation channels in IMCD cells. Am J Physiol 267: F558-F565.
    • Panten U, Bwgfeld J, Goerke F, Rennicke M, Schwanstecher M, Wallasch A, Zunkler B J, Lenzen S (1989) Control of insulin secretion by sulfonylureas, meglitinide and diazoxide in relation to their binding to the sulfonylurea receptor in pancreatic islets. Biochem Pharmacol 38: 1217-1229.
    • Perillan P R, Chen M, Potts E A, Simard J M (2002) Transforming growth factor-b 1 regulates Kir2.3 inward rectifier K+ channel via phospholipase C and protein kinase C-d in reactive astrocytes from adult rat brain. J. Biol. Chem. 277: 1974-1980.
    • Perillan P R, Li X, Potts E A, Chen M, Bredt D S, Simard J M (2000) Inward rectifier K+ channel Kir2.3 (W) in reactive astrocytes from adult rat brain. Glia 3 1: 181-192.
    • Perillan P R, Li X, Simard J M (1999) K+ inward rectifier currents in reactive astrocytes from adult rat brain. Glia 27: 213-225.
    • Popp R, Gogelein H (1992) A calcium and ATP sensitive nonselective cation channel in the antiluminal membrane of rat cerebral capillary endothelial cells. Biochim Biophys Acta 1108: 59-66.
    • Rae J L, Dewey J, Cooper K, Gates P (1990) A non-selective cation channel in rabbit corneal endothelium activated by internal calcium and inhibited by internal ATP. Exp Eye Res 50: 373-384.
    • Ransom C B, Sontheimer H (1995) Biophysical and pharmacological characterization of inwardly rectifying K+ currents in rat spinal cord astrocytes. J Neurophysiol 73: 333-346.
    • Renkin E M (1955) Filtration, diffusion, and molecular sieving through porous cellulose membranes. J Gen Physiol 38: 225-243.
    • Robinson R A, Stokes R H (1970) Electrolyte Solutions. London: Buttenvorths.
    • Rose C R, Waxman S G, Ransom B R (1998) Effects of glucose deprivation, chemical hypoxia, and simulated ischemia on Na+ homeostasis in rat spinal cord astrocytes. J Neurosci 18: 3554-3562.
    • Rucker-Martin C, Henaff M, Hatem S N, Delpy E, Mercadier J J (1999) Early redistribution of plasma membrane phosphatidylserine during apoptosis of adult rat ventricular myocytes in vitro. Basic Res Cardiol 94: 171-179.
    • Rutledge E M, Kimelberg H K (1996) Release of [3H]-D-aspartate from primary astrocyte cultures in response to raised external potassium. J Neurosci 16: 7803-78 1 1.
    • Schroder W, Hager G, Kouprijanova E, Weber M, Schmitt A B, Seifert G, Steirihauser C (1999) Lesion-induced changes of electrophysiological properties in astrocytes of the rat dentate gyrus. Glia 28: 166-1 74.
    • Shyng S, Ferrigni T, Nichols C G (1997) Control of rectification and gating of cloned KATP channels by the Kir6.2 subunit. J Gen Physiol 110: 141-153.
    • Sigworth F J, Sine S M (1987) Data transformations for improved display and fitting of single-channel dwell time histograms. Biophys J 52: 1047-1 054.
    • Staub F, Peters J, Kempski O, Schneider G H, Schurer L, Baethmann A (1993) Swelling of glial cells in lactacidosis and by glutamate: significance of C1-transport. Brain Res 610: 69-74.
    • Sturgess N C, Hales C N, Ashford M L (1987) Calcium and ATP regulate the activity of a non-selective cation channel in a rat insulinoma cell line. Pflugers Arch 409: 607-6 1 5.
    • Swanson R A (1992) Astrocyte glutamate uptake during chemical hypoxia in vitro. Neurosci Lett 147: 143-146.
    • Tanaka S, Uehara T, Nomura Y (2000) Up-regulation of protein-disulfide isomerase in response to hypoxiahrain ischemia and its protective effect against apoptotic cell death. J Biol Chem 275: 10388-10393.
    • Ubl J, Murer H, Kolb H A (1988) Ion channels activated by osmotic and mechanical stress in membranes of opossum kidney cells. J Membr Biol 104: 223-232.
    • Walz W, Gimpl G, Ohlemeyer C, Kettenmann H (1994) Extracellular ATP-induced currents in astrocytes: involvement of a cation channel. J Neurosci Res 38: 12-18.
    • Yu A C, Wong H K, Yung H W, Lau L T (2001) Ischemia-induced apoptosis in primary-cultures of astrocytes. Glia 35: 121-130.

Claims (43)

1. A composition comprising a membrane preparation expressing the NCCa-ATP channel.
2. The composition of claim 1, wherein said membrane preparation is derived from neural cells.
3. The composition of claim 1, wherein said membrane preparation is derived from isolated native reactive astrocytes (NRAs).
4. The composition of claim 1, wherein said channel is blocked by antagonists of the type 1 sulfonylurea receptor and opened by SUR1 activators.
5. The composition of claim 3, wherein said astrocytes are freshly isolated native reactive astrocytes.
6. The composition of claim 1, wherein said channel has the following characteristics: (a) it is a 35 pS type channel; (b) it is stimulated by cytoplasmic Ca2+ in the concentration range from about 10−8 to about 10−5 M; (c) it opens when cytoplasmic ATP is less than about 0.8 uM; and (d) it is permeable to the monovalent cations K+, Cs+, Li+ and Na+.
7. A method of screening for antagonists of the NCCa-ATP channel, comprising:
(a) contacting a test compound with a composition comprising the NCCa-ATP channel; and
(b) identifying test compounds that inhibit an activity of said channel by measuring said activity in the presence and absence of said test compound, wherein a test compound that inhibits said activity is identified as an antagonist of the NCCa-ATP channel.
8. The method of claim 7, wherein said composition comprises a preparation of neural cells expressing the NCCa-ATP channel.
9. The method of claim 7, wherein said composition comprises a membrane preparation expressing the NCCa-ATP channel.
10. The method of claim 7, wherein said membrane preparation is derived from isolated native reactive astrocytes (NRAs).
11. The method of claim 7, wherein said antagonist of the NCCa-ATP channel is selected from the group consisting of: (a) an NCCa-ATP channel inhibitor, an NCCa-ATP channel blocker, a SUR1 antagoinist, SUR1 inhibitor, and/or a compound capable of reducing the magnitude of membrane current through the channel.
12. A method for identifying compounds that inhibit neural cell swelling, comprising:
(a) contacting a test compound with a composition comprising the NCCa-ATP channel, and
(b) determining whether the test compound blocks the NCCa-Ap channel, wherein a test compound that blocks the NCCa-ATP channel is identified as a compound for inhibiting neural cell swelling.
13. A method for identifying compounds that inhibit brain swelling, comprising: (a) contacting a test compound with a composition comprising the NCCa-ATP channel, and (b) determining whether the test compound blocks the NCCa-ATP channel, wherein a test compound that blocks the NCCa-ATP channel is identified as a compound for inhibiting brain swelling.
14. A method for identifying compounds that inhibit brain swelling, comprising: (a) contacting a test compound with a composition comprising the NCCa-ATP channel, and (b) determining whether the test compound inhibits neural cell swelling, wherein a test compound that inhibits neural cell swelling is identified as a compound for inhibiting brain swelling.
15. A method for identifying compounds that inhibit neural cell swelling in an animal, comprising: (a) contacting a test compound with a composition comprising the NCCa-ATP channel and determining whether the test compound blocks the channel, and (b) administering the test compound to an animal having a brain injury or cerebral ischemia, and determining whether the test compound inhibits brain swelling of the treated animal, wherein test compounds that inhibit brain swelling are identified as compounds that inhibit neural cell swelling in an animal.
16. A method for identifying compounds that inhibit brain swelling, comprising:
(a) contacting a test compound with a composition comprising the NCCa-ATP channel, and determining whether the test compound blocks the channel, and
(b) administering the test compound to an animal having a brain injury or cerebral ischemia, and determining whether the test compound inhibits brain swelling of the treated animal, wherein test compounds that block the NCCa-ATP channel are identified as compounds that inhibit brain swelling.
17. The method according to any one of claims 12, 13, 14, 15 and 16, wherein said composition comprises a preparation of neural cells expressing the NCCa-ATP channel.
18. The method according to any one of claims 12, 13, 14, 15 and 16, wherein said composition comprises a membrane preparation expressing the NCCa-ATP channel.
19. The method according to any one of claims 12, 13, 14, 15 and 16, wherein said composition comprises a membrane preparation expressing the NCCa-ATP channel and further wherein said membrane preparation is derived from isolated native reactive astrocytes (NRAs).
20. The method according to any one of claims 12, 13, 14, 15 and 16, wherein said compound is an antagonist of a type 1 sulfonylurea receptor.
21. The method according to any one of claims 12, 13, 14, 15 and 16, wherein said compound is an antagonist of a type 1 sulfonylurea receptor, and further wherein said antagonist is a sulfonylurea compound.
22. The method according to any one of claims 12, 13, 14, 15 and 16, wherein said determining step is selected from the group consisting of:
(a) measuring channel currents;
(b) measuring membrane potential;
(c) detecting expression of annexin V;
(d) detecting expression of propidium iodide;
(e) in vitro binding assays;
(f) identifying appearance of cell blebbing and/or cell swelling; and
(g) combinations thereof.
23. The method according to any one of claims 12, 13, 14, 15 and 16, wherein said composition comprises a preparation of neural cells expressing the NCCa-ATP channel, and further wherein said cell is a native reactive astrocyte.
24. A method of preventing neural cell swelling in the brain of a subject, said method comprising administering to the subject a formulation containing an effective amount of a compound that blocks the NCCa-ATP channel and a pharmaceutically acceptable carrier.
25. A method of alleviating the negative effects of traumatic brain injury or cerebral ischemia stemming from neural cell swelling in a subject, comprising administering to the subject a formulation comprising an effective amount of a compound that blocks the NCCa-ATP channel and a pharmaceutically acceptable carrier.
26. The method according to any one of claims 24 and 25, wherein said administration is delivery into the brain.
27. The method according to any one of claims 24 and 25, wherein said compound is an antagonist of a type 1 sulfonylurea receptor.
28. The method according to any one of claims 24 and 25, wherein said compound is an antagonist of a type 1 sulfonylurea receptor, and further wherein said antagonist is a sulfonylurea compound.
29. The method according to any one of claims 24 and 25, wherein said compound is an antagonist of a type 1 sulfonylurea receptor, and further wherein said antagonist is selected from the group consisting of glibenclamide and tolbutamide.
30. The method according to any one of claims 24 and 25, wherein said compound is an antagonist of a type 1 sulfonylurea receptor, and further wherein said antagonist is selected from the group consisting of repaglinide, nateglinide, meglitinide, midaglizole, LY397364, LY389382, gliclazide, glimepiride, MgADP, and combinations thereof.
31. The method according to any one of claims 24 and 25, wherein said pharmaceutically acceptable carrier renders said formulation suitable for administration, said administration selected from the group consisting of intravenous, subcutaneous, intramuscular, intracutaneous, intragastric and oral administration.
32. A formulation for preventing or inhibiting neural cell swelling in the brain of a subject, wherein said formulation comprises a compound that blocks the NCCa-ATP channel and a pharmaceutically acceptable carrier, wherein the quantity of said compound is at least 10 times greater than the quantity of said compound in formulations for treating diabetes.
33. A method for identifying compounds that inhibit neural cell swelling, comprising:
(a) contacting a test compound with a composition comprising the Kir2.3 channel,
(b) determining whether the test compound opens the Kir2.3 channel, wherein a test compound that opens the Kir2.3 channel is identified as a compound for inhibiting neural cell swelling.
34. A method for identifying compounds that inhibit brain swelling, comprising: (a) contacting a test compound with a composition comprising the Kir2.3 channel, and (b) determining whether the test compound opens the Kir2.3 channel, wherein a test compound that opens the Kir2.3 channel is identified as a compound for inhibiting brain swelling.
35. A method for identifying compounds that inhibit neural cell swelling and/or brain swelling in an animal, comprising: (a) contacting a test compound with a composition comprising the Kir2.3 channel, and (b) determining whether the test compound opens the Kir2.3 channel, wherein a test compound that opens the Kir2.3 channel is identified as a compound for inhibiting neural cell swelling and/or brain swelling in an animal.
36. A method for identifying compounds that prevent, inhibit and/or alleviate brain swelling in a subject, comprising:
(a) contacting a test compound with a composition comprising the Kir2.3 channel, and determining whether the test compound opens the Kir2.3 channel, and
(b) administering the test compound to a subject having a brain injury or cerebral ischemia, and determining whether the test compound prevents, inhibits and/or alleviates brain swelling in the subject, wherein test compounds that open the Kir2.3 channel are identified as compounds that inhibit brain swelling.
37. A method for identifying compounds that inhibit neural cell swelling in an animal, comprising:
(a) contacting a test compound with a composition comprising the Kir2.3 channel, and determining whether the test compound opens the Kir2.3 channel, and
(b) administering the test compound to an animal having a brain injury or cerebral ischemia, and determining whether the test compound inhibits brain swelling of the treated animal, wherein test compounds that inhibit brain swelling are identified as compounds that inhibit neural cell swelling in an animal.
38. A method of preventing neural cell swelling in the brain of a subject, said method comprising administering to the subject a formulation containing an effective amount of a compound that opens the Kir2.3 channel and a pharmaceutically acceptable carrier.
39. A method of alleviating the negative effects of traumatic brain injury or cerebral ischemia stemming from neural cell swelling in a subject, comprising administering to the subject a formulation comprising an effective amount of a compound that opens the Kir2.3 channel and a pharmaceutically acceptable carrier.
40. The method according to any one of claims 33, 34, 35, 36, 37, 38 and 39, wherein said compound is Tenidap.
41. The method according to any one of claims 33, 34, 35, 36, 37, 38 and 39, wherein said determining step is selected from the group consisting of:
(a) measuring channel currents;
(b) measuring membrane potential;
(c) detecting expression of annexin V;
(d) detecting expression of propidium iodide;
(e) in vitro binding assays;
(f) identifying appearance of cell blebbing and/or cell swelling; and
(g) combinations thereof.
42. A composition comprising freshly isolated native reactive astrocytes, wherein said astrocytes express the NCCa-ATP channel.
43. The composition of claim 42, wherein at least about 80% to about 100% of said astrocytes express the NCCa-ATP channel.
US13/483,824 2002-03-20 2012-05-30 Methods for treating neural cell swelling Abandoned US20120237449A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US13/483,824 US20120237449A1 (en) 2002-03-20 2012-05-30 Methods for treating neural cell swelling
US14/184,947 US9107932B2 (en) 2002-03-20 2014-02-20 Methods for treating neural cell swelling
US14/815,154 US20150338393A1 (en) 2002-03-20 2015-07-31 Methods for treating neural cell swelling
US15/644,450 US10533988B2 (en) 2002-03-20 2017-07-07 Methods for treating central or peripheral nervous system damage
US15/895,945 US20180172671A1 (en) 2002-03-20 2018-02-13 Methods for treating neural cell swelling

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US36593302P 2002-03-20 2002-03-20
US10/391,561 US20030215889A1 (en) 2002-03-20 2003-03-20 Non-selective cation channel in neural cells and methods for treating brain swelling
US11/099,332 US7285574B2 (en) 2002-03-20 2005-04-05 Methods for treating neural cell swelling
US11/857,547 US8318810B2 (en) 2002-03-20 2007-09-19 Methods for treating neural cell swelling
US13/483,824 US20120237449A1 (en) 2002-03-20 2012-05-30 Methods for treating neural cell swelling

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/857,547 Continuation US8318810B2 (en) 2002-03-20 2007-09-19 Methods for treating neural cell swelling

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/184,947 Continuation US9107932B2 (en) 2002-03-20 2014-02-20 Methods for treating neural cell swelling

Publications (1)

Publication Number Publication Date
US20120237449A1 true US20120237449A1 (en) 2012-09-20

Family

ID=28454728

Family Applications (8)

Application Number Title Priority Date Filing Date
US10/391,561 Abandoned US20030215889A1 (en) 2002-03-20 2003-03-20 Non-selective cation channel in neural cells and methods for treating brain swelling
US11/099,332 Expired - Lifetime US7285574B2 (en) 2002-03-20 2005-04-05 Methods for treating neural cell swelling
US11/857,547 Active 2024-08-11 US8318810B2 (en) 2002-03-20 2007-09-19 Methods for treating neural cell swelling
US13/483,824 Abandoned US20120237449A1 (en) 2002-03-20 2012-05-30 Methods for treating neural cell swelling
US14/184,947 Expired - Lifetime US9107932B2 (en) 2002-03-20 2014-02-20 Methods for treating neural cell swelling
US14/815,154 Abandoned US20150338393A1 (en) 2002-03-20 2015-07-31 Methods for treating neural cell swelling
US15/644,450 Expired - Fee Related US10533988B2 (en) 2002-03-20 2017-07-07 Methods for treating central or peripheral nervous system damage
US15/895,945 Abandoned US20180172671A1 (en) 2002-03-20 2018-02-13 Methods for treating neural cell swelling

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US10/391,561 Abandoned US20030215889A1 (en) 2002-03-20 2003-03-20 Non-selective cation channel in neural cells and methods for treating brain swelling
US11/099,332 Expired - Lifetime US7285574B2 (en) 2002-03-20 2005-04-05 Methods for treating neural cell swelling
US11/857,547 Active 2024-08-11 US8318810B2 (en) 2002-03-20 2007-09-19 Methods for treating neural cell swelling

Family Applications After (4)

Application Number Title Priority Date Filing Date
US14/184,947 Expired - Lifetime US9107932B2 (en) 2002-03-20 2014-02-20 Methods for treating neural cell swelling
US14/815,154 Abandoned US20150338393A1 (en) 2002-03-20 2015-07-31 Methods for treating neural cell swelling
US15/644,450 Expired - Fee Related US10533988B2 (en) 2002-03-20 2017-07-07 Methods for treating central or peripheral nervous system damage
US15/895,945 Abandoned US20180172671A1 (en) 2002-03-20 2018-02-13 Methods for treating neural cell swelling

Country Status (12)

Country Link
US (8) US20030215889A1 (en)
EP (2) EP2438913B1 (en)
JP (1) JP4485806B2 (en)
AU (3) AU2003222020B2 (en)
CA (1) CA2477812C (en)
CY (1) CY1123341T1 (en)
DK (2) DK1529058T3 (en)
ES (2) ES2807274T3 (en)
HU (1) HUE051368T2 (en)
PT (1) PT2438913T (en)
SI (1) SI2438913T1 (en)
WO (1) WO2003079987A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060276411A1 (en) * 2002-03-20 2006-12-07 University Of Maryland, Baltimore Novel non-selective cation channel in neuronal cells and methods for treating brain swelling
US20090130083A1 (en) * 2004-09-18 2009-05-21 University Of Maryland Therapeutic Agents Targeting the NCCA-ATP Channel and Methods of Use Thereof
US20100092469A1 (en) * 2007-02-09 2010-04-15 Simard J Marc Antagonists of a non-selective cation channel in neural cells
US20100143347A1 (en) * 2007-01-12 2010-06-10 The University Of Maryland, Baltimore Targeting ncca-atp channel for organ protection following ischemic episode
US9107932B2 (en) 2002-03-20 2015-08-18 University Of Maryland, Baltimore Methods for treating neural cell swelling
US9375438B2 (en) 2007-06-22 2016-06-28 University Of Maryland, Baltimore Inhibitors of NCCa-ATP channels for therapy

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1782815B1 (en) * 2004-06-23 2010-09-08 Neurotec Pharma, S.L. Compound for use in the diagnosis of cns acute damage
CA2571717A1 (en) * 2004-06-23 2006-01-05 Josette-Nicole Mahy Gehenne Compounds for the treatment of inflammation of the central nervous system
AU2005287090A1 (en) 2004-09-18 2006-03-30 Department Of Veterans Affairs Therapeutic agents targeting the NCca-ATP channel and methods of use thereof
WO2007081946A2 (en) * 2006-01-09 2007-07-19 University Of South Florida Method for the identification of drugs to treat stroke at delayed timepoints
WO2008046014A1 (en) 2006-10-12 2008-04-17 Remedy Pharmaceuticals, Inc. Treatment of alzheimer's disease using compounds that reduce the activity of non-selective ca++- activated atp-sensitive cation channels regulated by sur1 receptors
US10004703B2 (en) 2006-10-12 2018-06-26 Biogen Chesapeake Llc Treatment of alzheimer's disease using compounds that reduce the activity of non-selective CA++ activated ATP-sensitive cation channels regulated by SUR1 channels
JP5812318B2 (en) * 2007-12-04 2015-11-11 レメディー ファーマシューティカルズ, インコーポレイテッド Improved formulations and methods for lyophilization and lyophilizates provided thereby
US20110034560A1 (en) * 2008-01-29 2011-02-10 Sven Jacobson Liquid formulations of compounds active at sulfonylurea receptors
WO2010033560A2 (en) 2008-09-16 2010-03-25 University Of Maryland, Baltimore Sur1 inhibitors for therapy
CA3225438A1 (en) 2010-07-19 2012-01-26 Remedy Pharmaceuticals, Inc. Methods of intravenous adminstration of glyburide and other drugs
EP2609914A1 (en) 2011-12-29 2013-07-03 Universitätsklinikum Hamburg-Eppendorf Novel methods for treating or preventing neurodegeneration
EP2846787B1 (en) 2012-05-08 2018-08-29 Aeromics, Inc. Compounds for use in the treatment of aquaporin-mediated diseases
SG11201604153UA (en) 2013-11-06 2016-07-28 Aeromics Inc Novel formulations
EP3359162B1 (en) * 2015-10-07 2022-12-21 Biogen Chesapeake LLC Methods of treating injuries or conditions related to cns edema
WO2018204721A1 (en) 2017-05-05 2018-11-08 Nino Sorgente Methods and compositions for improving eye health
US11382881B2 (en) 2017-05-05 2022-07-12 Nino Sorgente Methods and compositions for diagnosing and treating glaucoma
US20220168325A1 (en) * 2019-03-25 2022-06-02 The University Of Vermont Methods to promote cerebral blood flow in the brain
US20230348371A1 (en) * 2020-07-17 2023-11-02 Shanghai Senhui Medicine Co., Ltd. Sulfonylurea derivative and medical uses thereof
RU2752280C1 (en) * 2020-11-23 2021-07-26 федеральное государственное бюджетное образовательное учреждение высшего образования "Ростовский государственный медицинский университет" Министерства здравоохранения Российской Федерации (ФГБОУ ВО РостГМУ Минздрава России) Method for surgical treatment of malignant ischemic stroke

Family Cites Families (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6219552A (en) * 1985-07-19 1987-01-28 Ono Pharmaceut Co Ltd 3-((4-aroyl)phenoxy(or phenylthio))cyclopentanecarboxilic acid derivative, production thereof and remedy for cerebral edema containing said derivative as active constituent
GB8809205D0 (en) * 1988-04-19 1988-05-25 Nye P C G Treatment of oedema
US5166162A (en) * 1990-03-02 1992-11-24 Adir Et Compagnie Pyridylsulfonylurea and pyridylsulfonylthiourea compounds
CA2044853C (en) * 1990-07-19 2004-11-09 Susan A. Greenfield Method for treating parkinson's disease employing an atp-sensitive potassium channel blocker
US5236932A (en) * 1990-07-19 1993-08-17 E. R. Squibb & Sons, Inc. Method for treating Parkinson's disease employing quinine
CA2044855A1 (en) 1990-07-20 1992-01-21 Kerry P. S. J. Murphy Method for treating insult to neurons prone to parkinson's degeneration employing an atp-sensitive potassium channel blocker
US5215985A (en) * 1990-07-20 1993-06-01 E. R. Squibb & Sons, Inc. Method for treating ischemic insult to neurons employing an ATP-sensitive potassium channel blocker
WO1992006705A1 (en) 1990-10-16 1992-04-30 The Children's Medical Center Corporation Heparin binding mitogen with homology to epidermal growth factor (egf)
US5916871A (en) 1992-04-27 1999-06-29 Kansas State University Research Foundation Inhibitory factor
US6350739B1 (en) 1999-08-11 2002-02-26 University Of Florida Resarch Foundation, Inc. Methods of prevention and treatment of ischemic damage
DE4430212A1 (en) * 1994-08-28 1996-02-29 Merck Patent Gmbh Ortho-substituted benzoic acid derivatives
US20010003751A1 (en) * 1995-02-22 2001-06-14 Terashita Zen-Ichi Pharmaceutical composition for treating transient ischemic attack
AUPN193095A0 (en) * 1995-03-24 1995-04-27 Polychip Pharmaceuticals Pty Ltd Potassium ion channel blockers
US5545656A (en) * 1995-04-05 1996-08-13 Pfizer Inc. 2-Oxidole-1-carboxamide pharmaceutical agents for the treatment of alzheimer's disease
JPH09208562A (en) 1996-01-26 1997-08-12 Ono Pharmaceut Co Ltd Nitrogen monoxide synthetase inhibitor
US7608262B2 (en) * 1996-02-16 2009-10-27 The Kennedy Institute Of Rheumatology Methods of preventing or treating thrombosis with tumor necrosis factor antagonists
US5856360A (en) 1996-05-03 1999-01-05 Children's Hospital Medical Center Pharmaceutical method for the treatment of severe blood loss and for the inhibition or treatment of hemorrhagic shock
US6043224A (en) * 1996-09-05 2000-03-28 The Massachusetts Institute Of Technology Compositions and methods for treatment of neurological disorders and neurodegenerative diseases
US6184248B1 (en) 1996-09-05 2001-02-06 Robert K. K. Lee Compositions and methods for treatment of neurological disorders and neurodegenerative diseases
US20020016443A1 (en) 1996-10-04 2002-02-07 Keay Susan K. Antiproliferative factor
US5962645A (en) 1996-10-04 1999-10-05 University Of Maryland Antiproliferative factor from patients with interstitial cystitis
AU8283898A (en) 1997-06-30 1999-01-25 University Of Maryland At Baltimore Heparin binding-epidermal growth factor in the diagnosis of interstitial cystitis
US6056977A (en) * 1997-10-15 2000-05-02 Edward Mendell Co., Inc. Once-a-day controlled release sulfonylurea formulation
US6679859B1 (en) * 1997-10-24 2004-01-20 Alliance Pharmaceutical Corp. Amelioration of ischemic damage using synthetic oxygen carriers
AU1690399A (en) * 1997-12-26 1999-07-19 Mochida Pharmaceutical Co., Ltd. Neovascularization inhibitor containing dienogest as the active ingredient
US6180671B1 (en) 1998-03-10 2001-01-30 Beth Israel Deaconess Medical Center, Inc. Methods for treating disorders in which docosahexaenoic acid (DHA) levels are affected
US6100047A (en) * 1999-04-07 2000-08-08 Zen Bio, Inc. Modulation of the sulfonylurea receptor and calcium in adipocytes for treatment of obesity/diabetes
EP1075271A4 (en) 1998-04-17 2002-09-25 Univ Maryland Method of treating interstitial cystitis with recombinant heparin-binding epidermal growth factor-like growth factor (hb-egf)
US6613785B2 (en) * 1998-07-21 2003-09-02 Smithkline Beecham Plc Use of glucose uptake enhancer for reducing post-ischemic injury of the heart
EP1124950B1 (en) 1998-10-26 2006-07-12 Avi Biopharma, Inc. Morpholino based p53 antisense oligonucleotide, and uses thereof
PL351898A1 (en) * 1999-04-06 2003-06-30 Sankyo Co O-substituted derivatives of carboxylic acids
JP2002544258A (en) * 1999-05-12 2002-12-24 ニューロサーチ、アクティーゼルスカブ Ion channel regulator
JP2003510325A (en) * 1999-09-30 2003-03-18 ニューロジェン・コーポレーション Certain alkylenediamine-substituted pyrazolo [1,5-a] -1,5-pyrimidines and pyrazolo [1,5-a] -1,3,5-triazines
CA2393083C (en) * 1999-12-23 2010-01-26 Novartis Ag Use of hypoglycemic agent for treating impaired glucose metabolism
EP1251840A2 (en) 2000-01-26 2002-10-30 Cedars-Sinai Medical Center Method for using potassium channel activation for delivering a medicant to an abnormal brain region and/or a malignant tumor
EP1145717B1 (en) * 2000-04-13 2004-05-12 Pfizer Products Inc. Synergistic effect of glyburide and milrinone
WO2001082916A2 (en) * 2000-05-03 2001-11-08 Tularik Inc. Combination therapeutic compositions and methods of use
KR100867760B1 (en) 2000-05-15 2008-11-10 소니 가부시끼 가이샤 Reproducing apparatus, reproducing method and recording medium
CN100339132C (en) 2000-05-15 2007-09-26 史密丝克莱恩比彻姆公司 Antithrombotic agents
US6627636B2 (en) * 2000-06-15 2003-09-30 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
DE10054481A1 (en) * 2000-11-03 2002-05-08 Aventis Pharma Gmbh Acylaminoalkyl-substituted benzenesulfonamide derivatives, their preparation, their use and pharmaceutical compositions containing them
ITMI20010450A1 (en) 2001-03-05 2002-09-05 Univ Ferrara PROPHARMACIES DERIVED FROM ASCORBIC ACID AT THE PASSAGE OF THE BLOOD-ENCEPHALIC BARRIER
US6561075B2 (en) 2001-05-09 2003-05-13 Delphi Technologies, Inc. Power booster with mechanical panic assist function
US6492339B1 (en) * 2001-05-23 2002-12-10 Insmed, Incorporated Compositions comprising D-chiro inositol and sulfonylureas and methods of treatment thereof
US20060194750A1 (en) 2001-12-31 2006-08-31 Shuster Samuel J Methods and materials for modulating trpc4
US20030171407A1 (en) 2002-03-07 2003-09-11 Upsher-Smith Laboratories, Inc. Composition for reducing blood glucose and cholesterol
US8980952B2 (en) * 2002-03-20 2015-03-17 University Of Maryland, Baltimore Methods for treating brain swelling with a compound that blocks a non-selective cation channel
EP2438913B1 (en) * 2002-03-20 2020-05-06 University of Maryland, Baltimore A non-selective cation channel in neural cells and compounds that block the channel for use in treating brain swelling
US20050009733A1 (en) 2003-04-22 2005-01-13 Pharmacia Corporation Compositions of a cyclooxygenase-2 selective inhibitor and a potassium ion channel modulator for the treatment of central nervous system damage
US7326706B2 (en) 2003-08-15 2008-02-05 Bristol-Myers Squibb Company Pyrazine modulators of cannabinoid receptors
WO2005041877A2 (en) 2003-10-29 2005-05-12 Children's Medical Center Corporation Method of inhibiting rejection following organ transplantation
EP1782815B1 (en) 2004-06-23 2010-09-08 Neurotec Pharma, S.L. Compound for use in the diagnosis of cns acute damage
CN101043891A (en) 2004-09-18 2007-09-26 巴尔的摩马里兰大学 Therapeutic agents targeting the nc ca-atp channel and methods of use thereof
AU2005287090A1 (en) 2004-09-18 2006-03-30 Department Of Veterans Affairs Therapeutic agents targeting the NCca-ATP channel and methods of use thereof
WO2007011595A2 (en) 2005-07-15 2007-01-25 Neuren Pharmaceuticals Limited Neural regeneration peptides and antioxidants protect neurons from degeneration
AU2006269994A1 (en) 2005-07-15 2007-01-25 Children's Medical Center Corporation Methods for treating and diagnosing complications of premature birth
MX2008006204A (en) 2005-11-11 2008-10-17 Aurogen Inc Method for treating disease or disorder of adult central nervous system associated with tissue shrinkage or atrophy by administration of insulin.
WO2008089103A2 (en) 2007-01-12 2008-07-24 University Of Maryland, Baltimore Targeting ncca-atp channel for organ protection following ischemic episode
WO2008098160A1 (en) 2007-02-09 2008-08-14 University Of Maryland, Baltimore Antagonists of a non-selective cation channel in neural cells
US8557867B2 (en) 2007-06-22 2013-10-15 The United States Of America As Represented By The Department Of Veterans Affairs Inhibitors of NCCa-ATP channels for therapy
US7787809B2 (en) 2007-09-10 2010-08-31 Kabushiki Kaisha Toshiba Image forming apparatus, transfer unit thereof, and method of shifting transfer rollers thereof
US7813201B2 (en) 2008-07-08 2010-10-12 Atmel Corporation Differential sense amplifier
WO2010033560A2 (en) 2008-09-16 2010-03-25 University Of Maryland, Baltimore Sur1 inhibitors for therapy

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060276411A1 (en) * 2002-03-20 2006-12-07 University Of Maryland, Baltimore Novel non-selective cation channel in neuronal cells and methods for treating brain swelling
US8980952B2 (en) 2002-03-20 2015-03-17 University Of Maryland, Baltimore Methods for treating brain swelling with a compound that blocks a non-selective cation channel
US9107932B2 (en) 2002-03-20 2015-08-18 University Of Maryland, Baltimore Methods for treating neural cell swelling
US10533988B2 (en) 2002-03-20 2020-01-14 University Of Maryland, Baltimore Methods for treating central or peripheral nervous system damage
US20090130083A1 (en) * 2004-09-18 2009-05-21 University Of Maryland Therapeutic Agents Targeting the NCCA-ATP Channel and Methods of Use Thereof
US10583094B2 (en) 2004-09-18 2020-03-10 University Of Maryland Therapeutic methods that target the NCCA-ATP channel
US20100143347A1 (en) * 2007-01-12 2010-06-10 The University Of Maryland, Baltimore Targeting ncca-atp channel for organ protection following ischemic episode
US9511075B2 (en) 2007-01-12 2016-12-06 The University Of Maryland, Baltimore Targeting NCCA-ATP channel for organ protection following ischemic episode
US10166244B2 (en) 2007-01-12 2019-01-01 University Of Maryland, Baltimore Targeting NCCA-ATP channel for organ protection following ischemic episode
US10898496B2 (en) 2007-01-12 2021-01-26 University Of Maryland, Baltimore Targeting NCCa-ATP channel for organ protection following ischemic episode
US20100092469A1 (en) * 2007-02-09 2010-04-15 Simard J Marc Antagonists of a non-selective cation channel in neural cells
US9375438B2 (en) 2007-06-22 2016-06-28 University Of Maryland, Baltimore Inhibitors of NCCa-ATP channels for therapy

Also Published As

Publication number Publication date
EP2438913A1 (en) 2012-04-11
CY1123341T1 (en) 2021-12-31
ES2436467T3 (en) 2014-01-02
EP2438913B1 (en) 2020-05-06
SI2438913T1 (en) 2020-10-30
EP1529058B1 (en) 2013-11-06
WO2003079987A2 (en) 2003-10-02
JP4485806B2 (en) 2010-06-23
AU2003222020B2 (en) 2008-08-28
AU2011201252A1 (en) 2011-04-07
HUE051368T2 (en) 2021-03-01
AU2008243265B2 (en) 2010-12-23
DK1529058T3 (en) 2013-12-02
CA2477812C (en) 2014-10-14
US20030215889A1 (en) 2003-11-20
AU2003222020A1 (en) 2003-10-08
JP2005534285A (en) 2005-11-17
EP1529058A2 (en) 2005-05-11
US9107932B2 (en) 2015-08-18
ES2807274T3 (en) 2021-02-22
US8318810B2 (en) 2012-11-27
AU2011201252B2 (en) 2012-12-06
US10533988B2 (en) 2020-01-14
US20180172671A1 (en) 2018-06-21
CA2477812A1 (en) 2003-10-02
US20170307593A1 (en) 2017-10-26
PT2438913T (en) 2020-05-27
DK2438913T3 (en) 2020-06-22
US20080139659A1 (en) 2008-06-12
AU2008243265A1 (en) 2008-12-04
US20140235564A1 (en) 2014-08-21
EP1529058A4 (en) 2008-07-30
WO2003079987A3 (en) 2005-03-10
US20150338393A1 (en) 2015-11-26
US20050181980A1 (en) 2005-08-18
US7285574B2 (en) 2007-10-23

Similar Documents

Publication Publication Date Title
US10533988B2 (en) Methods for treating central or peripheral nervous system damage
CA2643360C (en) A novel non-selective cation channel in neuronal cells and methods for treating brain swelling
Chen et al. Cell swelling and a nonselective cation channel regulated by internal Ca2+ and ATP in native reactive astrocytes from adult rat brain
Levin et al. Aquaporins and CFTR in ocular epithelial fluid transport
Farrugia et al. Potassium current in circular smooth muscle of human jejunum activated by fenamates
Thorn et al. Nonselective cation channels in exocrine gland cells
Boyle et al. Permeabilization by streptolysin-O reveals a role for calcium-dependent protein kinase C isoforms alpha and beta in the response of cultured cardiomyocytes to hyposmotic challenge
Miwa et al. In vivo activities of aldose reductase inhibitors having a 1-(arylsulfonyl) hydantoin structure
Melvin et al. Altered responses to agonists after chronic in vivo atropine administration in rat parotid acini
CA2282236A1 (en) The use of excitatory amino acid transporter inhibitors to protect against cns white matter injury
Pearson et al. Biophysical and pharmacological characteristics of Ca2+ channel currents in rat cerebellar granule neurones are dependent on internal solution
Graham Airway Epithelial Sodium Transport
Hoffman Role of the extracellular matrix in the biochemistry of synaptic plasticity

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UNITED STATES OF AMERICAN AS REPRESENTED BY TH

Free format text: ASSIGNMENT OF JOINT UNDIVIDED RIGHT, TITLE AND INTEREST;ASSIGNOR:UNIVERSITY OF MARYLAND, BALTIMORE;REEL/FRAME:028306/0937

Effective date: 20120319

AS Assignment

Owner name: THE UNITED STATES OF AMERICA AS REPRESENTED BY THE

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNEE NAME SHOULD READ: THE UNITED STATES OF AMERICA AS REPRESENTED BY THE DEPT. OF VETERANS AFFAIRS PREVIOUSLY RECORDED ON REEL 028306 FRAME 0937. ASSIGNOR(S) HEREBY CONFIRMS THE THE UNITED STATE OF AMERICAN AS REPRESENTED BY THE DEPT. OF VETERANS AFFAIRS;ASSIGNOR:UNIVERSITY OF MARYLAND, BALTIMORE;REEL/FRAME:028561/0731

Effective date: 20120319

AS Assignment

Owner name: UNIVERSITY OF MARYLAND, BALTIMORE, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SIMARD, J. MARC;CHEN, MINGKUI;SIGNING DATES FROM 20030605 TO 20030606;REEL/FRAME:029017/0076

STCB Information on status: application discontinuation

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