Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS FOR TREATING MEMORY IMPAIRMENT AND COGNITIVE DYSFUNCTION
Document Type and Number:
WIPO Patent Application WO/2019/162945
Kind Code:
A1
Abstract:
The invention provides methods for treating a subject afflicted with a memory impairment or a cognitive dysfunction. Additional methods for improving memory and memory related functions in healthy subjects are also provided. The Methods include administration of an inhibitor of Na/K-ATPase.

Inventors:
KAPHZAN HANOCH (IL)
Application Number:
PCT/IL2019/050207
Publication Date:
August 29, 2019
Filing Date:
February 24, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CARMEL HAIFA UNIV ECONOMIC CORPORATION LTD (IL)
International Classes:
A61K31/585
Other References:
KAPHZAN, H. ET AL.: "Alterations in intrinsic membrane properties and the axon initial segment in a mouse model of Angelman syndrome", JOURNAL OF NEUROSCIENCE, vol. 31, no. 48, 19 October 2011 (2011-10-19), pages 17637 - 17648, XP055633213
CHAKRABORTY, D. ET AL.: "Selective ligands for Na+/K+-ATPase ? isoforms differentially and cooperatively regulate excitability of pyramidal neurons in distinct brain regions", NEUROPHARMACOLOGY, vol. 117, 20 February 2017 (2017-02-20), pages 338 - 351, XP029970775, DOI: 10.1016/j.neuropharm.2017.02.016
See also references of EP 3755340A4
Attorney, Agent or Firm:
GEYRA, Assaf et al. (IL)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method for treating a subject afflicted with a memory impairment or a cognitive dysfunction, comprising administering to said subject an inhibitor of Na/K- ATPase, thereby treating a subject afflicted with a memory impairment or a cognitive dysfunction.

2. The method of claim 1, wherein said Na/K-ATPase is al-NaKa.

3. The method of any one of claims 1 and 2, wherein said subject is further afflicted with a neurological disorder.

4. The method of any one of claims 1 to 3, wherein said subject is afflicted with Angelman syndrome.

5. The method of any one of claims 1 to 4, wherein said memory impairment comprises aberrant hippocampus-dependent long-term memory.

6. The method of any one of claims 1 to 5, wherein said inhibitor is selected from the list consisting of: digitalis, gitoxigenin, digoxigenin, digoxin, digitoxigenin, digitoxin, dihydrodigoxin, strophanthins, convallatoxin, cymarine, acetylstrophanthidin, strophanthidin, ouabagenin, ouabain, dihydrooubain, neriifolin, proscillaridin, proscillaridin A, cinobufagen, cinobufatolin, resibufagen, marinobufagenin, norbufalin, bufanolide, bufalin, and their respective isomers, inotropes, congeners, analogs, aglycone moieties and metabolites,.

7. The method of any one of claims 1 to 6, wherein said inhibitor is marinobufagenin.

8. The method of any one of claims 1 to 7, wherein said marinobufagenin is administered in an amount from 0.5pg/Kg/day to lmg/Kg/day.

9. The method of claim any one of claims 7 and 8, wherein said marinobufagenin is administered in an amount from 5pg/Kg/day to 50pg/Kg/day.

10. The method of any one of claims 1 to 9, wherein the duration of treatment is 14 days.

11. A method for improving memory or a memory related function in a subject in need thereof, comprising administering to said subject an inhibitor of Na/K-ATPase of Na/K-ATPase, thereby improving memory or a memory related function in a subject in need thereof.

12. The method of claim 11, wherein said Na/K-ATPase is al-NaKa.

13. The method of any one of claims 11 and 12, wherein said memory related function is selected from: learning, reasoning, alertness, attention, concentration, language processing, and social learning.

14. The method of any one of claims 11 to 13, wherein said memory is short-term memory or long-term memory.

15. The method of any one of claims 11 to 14, wherein said inhibitor is marinobufagenin.

16. The method of claim 15, wherein said marinobufagenin is administered in an amount between 0.5 pg/Kg/day to 1 mg/Kg/day.

17. The method of any one of claims 15 and 16, wherein said marinobufagenin is administered in an amount of 5pg/Kg/day to 50pg/Kg/day.

18. The method of any one of claim 11 to 17, wherein the duration of treatment is 14 days.

Description:
METHODS FOR TREATING MEMORY IMPAIRMENT AND

COGNITIVE DYSFUNCTION

CROSS REFERENCE TO RELATED APPLICATIONS

[01] This application claims the benefit of priority to U.S. Provisional Patent Application No. 62/634,256 filed February 23, 2018, entitled "METHODS FOR TREATING MEMORY IMPAIRMENT AND COGNITIVE DYSFUNCTION", the contents of which are incorporated herein by reference in their entirety.

FIELD OF INVENTION

[02] Present embodiments relate to the treatment of memory impairment and cognitive dysfunction including methods for treating memory impairments and cognitive dysfunctions.

BACKGROUND OF THE INVENTION

[03] Angelman syndrome (AS) is a human neuropsychiatric disorder associated with symptoms that include autism, mental retardation, motor abnormalities, epilepsy and lack of speech (Lossie et al., 2001; Williams et al., 2006). In most cases, AS is caused by the deletion of small portions on the maternal copy of chromosome 15, which includes the UBE3A gene (Kishino et al., 1997; Knoll et al., 1989; Matsuura et al., 1997; Sutcliffe et al., 1997). This phenomenon that only the maternal copy is expressed and the paternal allele is silenced, is called“imprinting”, and in AS it occurs mainly in the brain (Gustin et al., 2010). The UBE3A gene encodes an enzyme termed ubiquitin ligase E3A (also termed E6-AP), which is one of a family of enzymes that covalently attaches polyubiquitin chains to proteins, to signal for their degradation by the 26S proteasome. The prevalence of AS is 1/10,000-1/12,000 births (Petersen et al., 1995; Steffenburg et al., 1996) and it constitutes about ~2-5% of all children with established developmental delay (Buckley et al., 1998; Jacobsen et al., 1998).

[04] A mouse model of AS with UBE3A deletion has been generated and these mice exhibit susceptibility to epilepsy, impaired motor functioning, and cognitive abnormalities that correlate with neurological alterations observed in humans (Jiang et al., 1998). Like in other cases of autism (Bachevalier, 1994; DeLong, 1992; Dickerson and Eichenbaum, 2010; Sumiyoshi et ah, 2011), one of the main brain regions that is implicated in AS in humans and in the mouse model is the hippocampus (Jiang et ah, 1998). Hippocampus-dependent long-term memory (LTM) is impaired in these mice, and so is the cellular correlate of synaptic plasticity of hippocampal CA1 long-term potentiation in response to high frequency stimulation (HFS-LTP) (Jiang et al., 1998; van Woerden et al., 2007).

[05] Hippocampal CA1 and CA3 pyramidal cells from AS mouse model exhibit increased expression of alpha l-Na/K-ATPase (al-NaKA) and two axon initial segment (AIS) related proteins: NaVl.6 and ankyrin-G (Kaphzan et al., 2011).

[06] Prevention of al-NaKA increased expression in the hippocampus by genetic intervention rescues AS mice behavioral and electrophysiological hippocampal deficits (Kaphzan et al., 2013).

SUMMARY OF THE INVENTION

[07] In one embodiment, the present invention provides a method for treating a subject afflicted with a memory impairment or a cognitive dysfunction, comprising administering to said subject an inhibitor of Na/K-ATPase, thereby treating a subject afflicted with a memory impairment or a cognitive dysfunction.

[08] In another embodiment, the present invention provides a method for treating a subject afflicted with a memory impairment or a cognitive dysfunction, comprising administering to said subject an inhibitor of al-NaKa, thereby treating a subject afflicted with a memory impairment or a cognitive dysfunction.

[09] In another embodiment, the present invention provides a method for treating a subject afflicted with a memory impairment or a cognitive dysfunction, wherein the subject is further afflicted with a neurological disorder.

[010] In another embodiment, the present invention provides a method for treating a subject afflicted with a memory impairment or a cognitive dysfunction, wherein the subject is afflicted with Angelman syndrome. [011] In another embodiment, the present invention provides a method for treating a subject afflicted with a memory impairment or a cognitive dysfunction, wherein the memory impairment comprises aberrant hippocampus-dependent long-term memory.

[012] In another embodiment, the present invention provides a method for treating a subject afflicted with a memory impairment or a cognitive dysfunction, comprising administering to said subject an inhibitor of Na/K-ATPase, wherein the Na/K-ATPase inhibitor is selected from the list consisting of: digitalis, gitoxigenin, digoxigenin, digoxin, digitoxigenin, digitoxin, dihydrodigoxin, strophanthins, convallatoxin, cymarine, acetylstrophanthidin, strophanthidin, ouabagenin, ouabain, dihydrooubain, neriifolin, proscillaridin, proscillaridin A, cinobufagen, cinobufatolin, resibufagen, marinobufagenin, norbufalin, bufanolide, bufalin, and their respective isomers, inotropes, congeners, analogs, aglycone and metabolites.

[013] In another embodiment, the present invention provides a method for treating a subject afflicted with a memory impairment or a cognitive dysfunction, comprising administering to said subject an inhibitor of Na/K-ATPase, wherein the Na/K-ATPase inhibitor is marinobufagenin.

[014] In another embodiment, the method comprises administration of marinobufagenin in an amount from 0.5pg/Kg/day to lmg/Kg/day.

[015] In another embodiment, the method comprises administration of marinobufagenin in an amount from 5pg/Kg/day to 50pg/Kg/day.

[016] In another embodiment, the duration of treatment is 14 days.

[017] In another aspect, the present invention provides a method for improving memory or a memory related function in a subject in need thereof, comprising administering to said subject an inhibitor of Na/K-ATPase of Na/K-ATPase, thereby improving memory or a memory related function in a subject in need thereof.

[018] In another embodiment, the present invention provides a method for improving memory or a memory related function in a subject in need thereof, wherein the memory related function is selected from: learning, reasoning, alertness, attention, concentration, language processing, and social learning. [019] In another embodiment, the present invention provides a method for improving memory or a memory related function in a subject in need thereof wherein memory is short-term memory or long-term memory.

BRIEF DESCRIPTION OF THE DRAWINGS

[020] Figure 1 is a graph showing correction of long-term potentiation defects in AS mice by administration of 0.5mM marinobufagenin.

[021] Figure 2 is a graph showing the effect of administration of IOmM marinobufagenin on resting membrane potential of CA1 pyramidal cells from the hippocampus of AS compared to WT mice.

DETAILED DESCRIPTION OF THE INVENTION

[022] In one embodiment, provided a method for treating a subject with a memory impairment or a cognitive dysfunction comprising administering to the subject an agent that inhibits the activity of Na/K-ATPase.

[023] In another embodiment, the term "subject" refers to a human having a condition of impairment of memory or cognitive dysfunction. In another embodiment, the term "subject" refers to a mammal such as a pet or a farm animal.

[024] In another embodiment, the term "memory impairment" refers to a reduced ability to generate and retain memory. In another embodiment, the term "memory impairment" refers to a loss of existing memory. In another embodiment, the term "memory impairment" refers to a below average capacity to store memory for a short term and/or long term.

[025] In another embodiment, the method of the present invention is used to improve a memory impairment in a subject in need thereof. In another embodiment, the improvement in memory is an improvement in at least one result of a memory assessment test. Memory assessment tests using methodologies from neuropsychology, human development and animal testing are well known in the art. Non-limiting examples of such tests are: visual short-term memory (VSTM), spatial working memory (SPWM), stroop task, attention blink, task switch, flanker task, visual search task, perceptual motor speed (PMS), digit Span and basic processing Speed.

[026] In another embodiment, the term "cognitive dysfunction" as used herein, refers to a reduced ability of a subject to perceive, remember and act on information in the environment. In another embodiment, the method of the present invention is used to enhance at least one cognitive function in a subject in need thereof. In another embodiment, the enhancement of cognitive function is an improvement in at least one result of a cognitive function assessment test. Methods for cognitive function assessment of humans and other animals are well known in the art. Non-limiting examples of such methods are: inductive reasoning tests, inductive reasoning aptitude, intelligence quotient, situational judgement test, intelligence tests, Kohs block, Miller analogies test, Otis-Lennon School ability test, Raven's progressive matrices, Stanford-Binet IQ test, Wechsler adult intelligence scale, Wechsler intelligence scale for children, Wechsler preschool and primary scale of intelligence, wonderlic test, Cambridge neuropsychological test automated battery, CDR computerized assessment system, cognitive bias, cognitive pretesting, cognitive process profile (CPP),draw-a- person test, knox cubes, modern language aptitude test, Pimsleur language aptitude battery, porteus maze test, consensus based assessment, knowledge organization, knowledge hierarchies, and knowledge Ontologies.

[027] In another embodiment, memory improvement and/or cognitive function enhancement may be assessed by neuroscientific tools and methods. Non-limiting examples of such tools and methods are: functional MRI (fMRI), repetitive transcranial magnetic stimulation (rTMS), magnetoencephalography (MEG), positron emission tomography (PET) and electroencephalography (EEG).In another embodiment, the method of the present invention improves the results of a neuroscientific experiment of the subject as compared to pretreatment results.

[028] In another embodiment, the method of the present invention is used for improving short-term memory in a subject in need thereof. The term "short-term memory" is known in the art as brief retention of information lasting from minutes to a few hours. In some embodiments, the improvement is an improvement of 5%-80% of short-term memory. In some embodiments, the improvement is an improvement of 5%-50% of short-term memory. In some embodiments, the improvement is an improvement of 5%-30% of short-term memory. In some embodiments, the improvement is an improvement of 5%-15% of short-term memory. In some embodiments, the improvement is an improvement of 30%-80% of short-term memory. In some embodiments, the improvement is an improvement of 50%-80% of short-term memory.

[029] In another embodiment, the method of the present invention is used for improving long-term memory in a subject in need thereof. The term "long-term memory" is known in the art as the ability to retain information for long periods of days to years. In some embodiments, the improvement is an improvement of 5%-80% of long-term memory. In some embodiments, the improvement is an improvement of 5%-50% of long-term memory. In some embodiments, the improvement is an improvement of 5%-30% of long-term memory. In some embodiments, the improvement is an improvement of 30%-80% of long-term memory. In some embodiments, the improvement is an improvement of 50%-80% of long-term memory. In some embodiments, the improvement is an improvement of 5% -15% of long-term memory.

[030] In another embodiment, the method of the present invention is used for enhancing a cognitive function in a subject in need thereof. In some embodiments, the enhancement is an enhancement of at least one cognitive function of 5% -80%. In some embodiments, the enhancement is an enhancement of at least one cognitive function of 5%-50%. In some embodiments, the enhancement is an enhancement of at least one cognitive function of 30%-80%. In some embodiments, the enhancement is an enhancement of at least one cognitive function of 50% -80%. In some embodiments, the enhancement is an enhancement of at least one cognitive function of 5%-30%. In some embodiments, the enhancement is an enhancement of at least one cognitive function of 5%- 15% .

[031] In another embodiment, the term "improvement" as used herein means an increment in the result of at least one memory assessment test when compared to pre treatment results of the subject. In another embodiment, the improvement is compared to reference data. In another embodiment, the term "improvement" comprises enabling prolonging memory of a patient in need thereof. In another embodiment, the term "improvement" comprises enhancing the ability to memorize. In another embodiment, the term "improvement" comprises enhancing memory capacity. [032] In another embodiment, the term "enhancement" as used herein means an increment in the result of at least one cognitive function assessment test when compared to pre-treatment results of the subject. In another embodiment, the enhancement is compared to reference data.

[033] The term "reference data" as used herein refers to results of memory tests and/or cognitive function tests of a defined population which serves as a standard for assessing the improvement of the treated subject.

[034] In another embodiment, the method of the present invention is used in the treatment of diseases and/or pathologies that include and/or involve a memory impairment and/or a cognitive dysfunction. In another embodiment, the method of the present invention improves a memory impairment and/or a cognitive dysfunction in a subject suffering from a neurological condition. In another embodiment, the method of the present invention improves a memory impairment and/or a cognitive dysfunction in a subject suffering from a brain infection. In another embodiment, the method of the present invention improves a memory impairment and/or a cognitive dysfunction in a subject suffering from nutritional deficiencies. In another embodiment, the method of the present invention improves a memory impairment and/or a cognitive dysfunction in a subject suffering from age-associated memory impairment. In another embodiment, the method of the present invention improves a memory impairment and/or a cognitive dysfunction in a subject suffering from injuries of the brain. In another embodiment, the method of the present invention improves a memory impairment and/or a cognitive dysfunction in a subject suffering from adverse effects of a treatment to a medical condition such as, but not limited to, chemotherapy or radiotherapy for cancer treatment. In another embodiment, the method of the present invention improves a memory impairment and/or a cognitive dysfunction in a subject suffering a cognitive disorder.

[035] In another embodiment, the method of the present invention inhibits deterioration in memory capacity. In another embodiment, the method of the present invention inhibits deterioration in memory duration. Deterioration refers to a decline in a result of at least one memory assessment test of the subject as compared to a previous measurement. In one embodiment, a subject susceptible to a memory impairment is treated according to the present methods. In another embodiment, a subject susceptible to a memory impairment is a subject afflicted with a neurological disorder such as Alzheimer's disease. In another embodiment, a subject susceptible to a memory impairment is a subject afflicted with a neurological disorder such as Angelman syndrome. In another embodiment, a subject susceptible to a memory impairment is a subject afflicted with a neurological disorder such as autism spectrum disorders. In another embodiment, a subject susceptible to a memory impairment is a subject afflicted with a neurological disorder such as Rett syndrome. In another embodiment, a subject susceptible to a memory impairment is a subject afflicted with a neurological disorder such as Huntington's disease.

[036] In another embodiment, the term "treating” may include preventing, inhibiting or relieving a memory impairment and/or a cognitive dysfunction in a subject in need thereof by administering compounds as described herein.

[037] In another embodiment, preventing relates to a subject that may be predisposed to memory impairment and/or cognitive dysfunction but does not yet experience or exhibit such symptoms.

[038] In another embodiment, inhibiting memory impairment and/or a cognitive dysfunction refers to slowing or arresting their development in a subject exhibiting memory impairment and/or a cognitive dysfunction.

[039] In another embodiment, relieving memory impairment or cognitive dysfunction in a subject having these symptoms refers to causing regression of the symptoms as compared to a previous measure of the subject. In some embodiments, the regression may be of 5%-80%. In some embodiments, the regression may be of 50%-80%. In some embodiments, the regression may be of 30%-80%. In some embodiments, the regression may be of 5%-50%. In some embodiments, the regression may be of 5%-30%.In some embodiments, the regression may be of 5% -15%.

[040] In another embodiment, the term "administering" as used herein, includes delivery of a composition or one or more pharmaceutically active ingredients to a subject, by any appropriate methods, which serve to deliver the composition or its active ingredients or other pharmaceutically active ingredients to the subject. In another embodiment, the method of administration may vary depending on various factors, such as for example, the components of the pharmaceutical composition or the nature of the pharmaceutically active or inert ingredients, the site of the potential or actual malady, age and physical condition of the subject. Some non-limiting examples of ways to administer a composition or a pharmaceutically active ingredient to a subject include: oral, intravenous, topical, intrarespiratory, intraperitoneal, intramuscular, parenteral, sublingual, transdermal, intranasal, aerosol, intraocular, intratracheal, intrarectal, vaginal, gene gun, dermal patch, eye drop, ear drop or mouthwash.

[041] In another embodiment, the term "Na+/K+-ATPase" (sodium-potassium adenosine triphosphatase) as used herein and in the art, is an enzyme found in the plasma membrane of all animal cells. The Na-i- /K+ATPase enzyme is a solute pump that pumps sodium out of cells while pumping potassium into cells, against their concentration gradients. In another embodiment, compounds of the invention inhibit the activity of Na+/K+-ATPase. In another embodiment, compounds of the invention inhibit the amount of Na+/K+-ATPase. In another embodiment, compounds of the invention inhibit directly and/or indirectly the activity of Na+/K+-ATPase.

[042] In another embodiment, the term "inhibitor" refers to any molecule which is capable of preventing or reducing the biochemical activity of Na+/K+-ATPase. Without being bound to any particular theory, the inhibition is achieved by a mechanism selected from: physical binding of the inhibitor to the target molecule thereby interfering with the activity of the target molecule, competing with the binding of substrates of the target molecule, preventing or reducing the expression of a protein target, interfering with the sub-cellular localization of the target molecule. In another embodiment, the Na+/K+-ATPase inhibitor is a competitive inhibitor. In another embodiment, the Na+/K+-ATPase inhibitor is a non-competitive inhibitor. In another embodiment, the Na+/K+-ATPase inhibitor is a selective inhibitor. In another embodiment, the Na+/K+-ATPase inhibitor is a non-selective inhibitor.

[043] In another embodiment, the method comprises administering to the subject one or more compounds that inhibit Na+/K+ ATPase such as, but not limited to, "Cardiotonic steroids" (CTS). CTS include cardenolide or bufadienolide, or combination thereof. Non-limiting examples of cardenolides and bufadienolides are: digitalis, gitoxigenin, digoxigenin, digoxin, digitoxigenin, digitoxin, dihydrodigoxin, strophanthins, convallatoxin, cymarine, acetylstrophanthidin, strophanthidin, ouabagenin, ouabain, dihydrooubain, neriifolin, proscillaridin, proscillaridin A, cinobufagen, cinobufatolin, resibufagen, marinobufagenin, norbufalin, bufanolide, bufalin and similar compounds, and their respective isomers, inotropes, congeners, analogs, aglycone moieties and other variants, derivatives, equivalents, precursors and metabolites, and combination thereof.

[044] In another embodiment, the term "al-NaKa" (Sodium/potassium transporting ATPase subunit alpha- 1) as used herein, refers to an enzyme that in humans is encoded by the ATP1A1 gene (genebank accession number: NP_000692.2). Na+/K+- ATPase is composed of two subunits, a large catalytic subunit (alpha) and a smaller glycoprotein subunit (beta). There are four isoforms of the alpha subunit: a 1-4. The al isoform is the common isoform that maintains Na and K gradients in all tissues.

[045] In another embodiment, the method comprises administering to the subject a selective inhibitor of the al isoform of Na, K- ATPase. The term "selective inhibitor of the al isoform of Na, K- ATPase" means that the compound inhibits the al isoform of Na, K-ATPase to a greater degree than the other isoforms. In some embodiments, the selectivity of the compound for the al isoform of Na, K-ATPase is up to about 20 fold over other isoforms, e.g., up to 16 fold, 8 fold, 5 fold or 2 fold greater inhibition of the al isoform over other isoforms of this enzyme. As a non-limiting example, ouabain and digoxin are a3 inhibitors while Marinobufagenin is an a 1 inhibitor.

[046] In another embodiment, the selective inhibitor of the al isoform of Na, K- ATPase is marinobufagenin (MBG). In another embodiment, the selective inhibitor of the al isoform of Na, K-ATPase is resibufogenin.

[047] In another embodiment, the subject suffering from memory impairment and/or a cognitive dysfunction is further afflicted with a neurological disorder. The term "neurological disorder" as used herein refers to any disorder of the nervous system that can result in a range of symptoms. According to some embodiment, the methods of the invention are used for treating a memory impairment and/or a cognitive dysfunction in a subject suffering from a neurological disorder. Non limiting examples of neurological disorders that are treatable by the methods disclosed herein are selected from: Angelman syndrome, Rett syndrome, autism spectrum disorders, Alzheimer's disease, Huntington's disease, Lewy Body disease, dementia, cerebral atrophy, frontotemporal lobar degeneration, Pick's disease, multi infarct dementia, Down's syndrome, neurotoxic injury, cerebral hypoxia/ischemia, traumatic brain injury, cholinergic hypofunction, vascular narrowing or blockage in the brain, decreased cerebral perfusion, neuroinflammation or cognitive disorders.

[048] In some embodiments, the neurological disorder is a cognitive disorder. The term "cognitive disorder" as used herein refers to a condition that leads to cognitive dysfunction. Non-limiting examples of cognitive disorders include, but are not limited to: amnesia, dementia, and delirium, anxiety disorders, mood disorders, psychotic disorders, temporary hypoxia of the brain due to various reasons and mild cognitive impairment.

[049] In another embodiment, the subject is afflicted with Angelman syndrome. The Term "Angelman syndrome" (AS) is known in the art as a genetic disorder that causes developmental disabilities and neurological problems, such as difficulty in speaking, balancing and walking, memory impairment and cognitive dysfunction. In another embodiment, the method is used to prevent memory impairment and/or a cognitive dysfunction in a subject afflicted with Angelman syndrome. In another embodiment, the method is used to inhibit memory impairment and/or a cognitive dysfunction in a subject afflicted with Angelman syndrome. In another embodiment, the method is used to relive memory impairment and/or a cognitive dysfunction in a subject afflicted with Angelman syndrome.

[050] As used herein "Long-term memory" is a process that enables the brain to store information for long periods of time. The formation of long-term memory depends on a cellular process known in the art as long-term potentiation (LTP). Defects in LTP in the hippocampus may cause aberrant hippocampus-dependent long term memory. In another embodiment, administration of compounds of the present invention increases hippocampal LTP, thereby improving hippocampus-dependent long-term memory in a subject in need thereof. In another embodiment, the increase in hippocampal LTP is in response to high frequency stimulation. The term "high frequency stimulation" as used herein refers to a biochemical assay for testing long term potentiation described in the materials and methods section below.

[051] In another embodiment, administration of 0.5pg/Kg/day to lmg/Kg/day MBG results in a memory increase. In another embodiment, administration of lpg/Kg/day to 500pg /Kg/day MBG results in a memory increase. In another embodiment, administration of 5mg/Kg/day to 50mg /Kg/day MBG results in a memory increase.

[052] In some embodiments, a memory increase is an increase of 5% to 80% in hippocampus -dependent long-term memory formation. In another embodiment, a memory increase is an increase of 5% to 30% in hippocampus-dependent long-term memory formation. In another embodiment, a memory increase is an increase of 20% to 60% in hippocampus-dependent long-term memory formation. In another embodiment, a memory increase is an increase of 30% to 80% in hippocampus- dependent long-term memory formation. In another embodiment, a memory increase is an increase of 5% to 15% in hippocampus-dependent long-term memory formation.

[053] In another embodiment, administration of Na/K-ATPase inhibitor increases LTP induction compared to base-line, wherein base line is determined as the LTP induction of an untreated or a mock treated subject as described in example 1. In some embodiments, an increase in LTP induction improves hippocampus-dependent long term memory formation in a subject in need thereof.

[054] In another embodiment, administration of Na/K-ATPase inhibitor increases neuronal excitability in the hippocampus of an AS afflicted subject thereby improving hippocampus -dependent long-term memory formation.

[055] In another embodiment, administration of Na/K-ATPase inhibitor enhances synaptic plasticity in the hippocampus of an AS afflicted subject thereby improving hippocampus -dependent long-term memory formation.

[056] In some embodiments, the method of the present invention is used for improving one or more membrane properties of pyramidal cells of the CA1 region in the hippocampus of an AS afflicted subject thereby improving hippocampus- dependent long-term memory formation of the subject. Methods for measuring membrane properties of CA1 pyramidal cells have been described by Kaphzan et al. 2013. In some embodiments, the method of the present invention improves at least one membrane property selected from: improvement in the resting membrane potential of CA1 pyramidal cells as described in example 2, improvement in membrane time constant, improvement in threshold potential, improvement in action potential amplitude and improvement in the maximal rise of action potential. [057] In another embodiment, the selective inhibitor of the al isoform of Na, K- ATPase is marinobufagenin (MBG) having the formula:

3 -beta, 5 -dihydroxy- 14,15 -beta-epoxy-5-beta-bufa-20,22- dienolide .

[058] In another embodiment, the duration of the treatment is between 24h to 14 days. In another embodiment, the duration of the treatment is between 24h to 30 days. In another embodiment, the duration of the treatment is between 1 to 12 months. In another embodiment, the method is used to treat a subject with a chronic need for improving a memory impairment or a cognitive dysfunction and the duration of the treatment is for the life time of the subject.

[059] In another embodiment, the subject in need is a healthy human in need of improving memory or a memory related function such as, but not limited to, learning, reasoning, alertness, attention, concentration, language processing, social learning, or any combination thereof.

[060] In another embodiment, the method is used to improve memory or a memory related function in a subject in need of improving learning skills. In another embodiment, the method is used to improve memory or a memory related function in a subject having to face ongoing mental fatigue. In another embodiment, the method is used to improve memory or a memory related function in a subject having to face mental daily stress. In another embodiment, the method is used to improve memory or a memory related function in a subject having to face occupational stress. In another embodiment, the method is used to improve memory or a memory related function in a subject having to face multi tasks activities. In another embodiment, the method is used to improve memory or a memory related function in a subject exhibiting memory loss caused by hormonal changes or imbalances. In another embodiment, the method is used to improve memory or a memory related function in a subject exhibiting transient memory loss. In another embodiment, the method is used to improve memory or a memory related function in a subject exhibiting age related memory loss.

[061] In another embodiment, the method of the present invention is used to improve at least one memory related function in a subject in need thereof as measured by the methods of the present invention. In another embodiment, the improvement of a memory related function refers to an increased ability to perform a memory related function as compared to pre-treatment levels of the subject. In another embodiment, improvement is measured as compared to reference data.

[062] As used herein, "learning" refers to an individual's ability of acquiring new knowledge, or modifying and reinforcing, long lost existing knowledge, behaviors, skills, values, or preferences and may involve synthesizing different types of information.

[063] As used herein, "reasoning" refers to the use of reason, especially to form conclusions, inferences, or judgments.

[064] As used herein, "alertness" refers to a state of active attention by high sensory awareness such as being watchful and prompt to meet danger or emergency, or being quick to perceive and act.

[065] As used herein, "attention" refers to an individual's ability to focus on information that is relevant to a task at hand while ignoring information that is not relevant to the particular task.

[066] As used herein, "concentration" refers to an individual's ability to direct one's thinking in whatever direction one would intend.

[067] As used herein, "language processing" refers to the way humans use words to communicate ideas and feelings, and how such communications are processed and understood.

[068] As used herein the term“social learning” relates to knowledge and skills obtained within a social context including for example observational learning, imitation, and modeling, and use of such information to serve as a guide for action on subsequent occasions.

[069] In another embodiment, administration of 0.5pg/Kg/day to lmg/Kg/day MBG results in improvement of memory or a memory related function. In another embodiment, administration of lpg/Kg/day to 500pg /Kg/day MBG results in improvement of memory or a memory related function. In another embodiment, administration of 5pg/Kg/day to 50pg /Kg/day MBG results in in improvement of memory or a memory related function.

[070] In some embodiments, an improvement of memory or a memory related function is an increase of 5% to 80% compared to pre-treatment levels of the subject. In some embodiments, an improvement of memory or a memory related function is an increase of 20% to 60% compared to pre-treatment levels of the subject. In some embodiments, an improvement of memory or a memory related function is an increase of 30% to 80% compared to pre-treatment levels of the subject. In some embodiments, an improvement of memory or a memory related function is an increase of 5% to 15% compared to pre-treatment levels of the subject.

EXAMPLES

[071] Generally, the nomenclature used herein and the laboratory procedures utilized in the present invention include molecular, biochemical, microbiological and recombinant DNA techniques. Such techniques are thoroughly explained in the literature. See, for example, "Molecular Cloning: A laboratory Manual" Sambrook et al., (1989); "Current Protocols in Molecular Biology" Volumes I-III Ausubel, R. M., ed. (1994); Ausubel et al., "Current Protocols in Molecular Biology", John Wiley and Sons, Baltimore, Maryland (1989); Perbal, "A Practical Guide to Molecular Cloning", John Wiley & Sons, New York (1988); Watson et al., "Recombinant DNA", Scientific American Books, New York; Birren et al. (eds) "Genome Analysis: A Laboratory Manual Series", Vols. 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies as set forth in U.S. Pat. Nos. 4,666,828; 4,683,202; 4,801,531; 5,192,659 and 5,272,057; "Cell Biology: A Laboratory Handbook", Volumes I-III Cellis, J. E., ed. (1994); "Culture of Animal Cells - A Manual of Basic Technique" by Freshney, Wiley-Liss, N. Y. (1994), Third Edition; "Current Protocols in Immunology" Volumes I-III Coligan J. E., ed. (1994); Stites et al. (eds), "Basic and Clinical Immunology" (8th Edition), Appleton & Lange, Norwalk, CT (1994); Mishell and Shiigi (eds), "Selected Methods in Cellular Immunology", W. H. Freeman and Co., New York (1980); available immunoassays are extensively described in the patent and scientific literature, see, for example, U.S. Pat. Nos. 3,791,932; 3,839,153; 3,850,752; 3,850,578; 3,853,987; 3,867,517; 3,879,262; 3,901,654; 3,935,074; 3,984,533; 3,996,345; 4,034,074; 4,098,876; 4,879,219; 5,011,771 and 5,281,521; "Oligonucleotide Synthesis" Gait, M. J., ed. (1984); “Nucleic Acid Hybridization" Hames, B. D., and Higgins S. J., eds. (1985); "Transcription and Translation" Hames, B. D., and Higgins S. J., eds. (1984); "Animal Cell Culture" Freshney, R. L, ed. (1986); "Immobilized Cells and Enzymes" IRL Press, (1986); "A Practical Guide to Molecular Cloning" Perbal, B., (1984) and "Methods in Enzymology" Vol. 1-317, Academic Press; "PCR Protocols: A Guide To Methods And Applications", Academic Press, San Diego, CA (1990); Marshak et ah, "Strategies for Protein Purification and Characterization - A Laboratory Course Manual" CSHL Press (1996); all of which are incorporated by reference. Other general references are provided throughout this document.

MATERIALS AND METHODS

[072] Animals: Mice for experiments were bred from a female that was heterozygous for the deletion of Ube3a from a paternal origin (Ube3a p-/m+) and an al-NaKA heterozygous male (al-NaKA p-/m+). Both parents were on a C57/B16 background. For all experiments mice used were 8-12 weeks of age (unless otherwise specified), and for all experiments littermates were used as controls. Mice were genotyped using specific primers as described previously (Jiang et ah, 1998) and (James et al., 1999).

[073] Electrophysiology: Hippocampal slices were prepared from wild-type and AS model mice as previously described (Kaphzan et al., 2012; Kaphzan et al., 2013). LTP was induced in area CA1 with high frequency stimulation (HFS) by delivering two 100 Hz trains of HFS with an intertrain interval of 20 sec. The initial slope of the field excitatory postsynaptic potential (fEPSP) for each experiment was expressed as the ratio to the average baseline response prior to delivery of HFS.

[074] Drug handling: Marinobufagenin was kept in stock solution (1 mg/lO ml) in 0.5% DMSO and was freshly diluted to the required concentration 0.042mg/mL by saline prior to the experiment. At this stock concentration a volume of lOOpL for 16 days in a rate of 0.25pL/hour correlated to approximately l0pg/kg/day for a mouse of 25 grams. The exact concentration was calculated for each mouse according to its body weight. As a vehicle 1:200 stock solution of DMSO in water (the maximal concentration of DMSO matched to the maximal dose of MBG, i.e. lOpg/kg/day) was used. For delivery via osmotic pumps only mice that weigh more than 20g were used. For LTP studies, marinobufagenin was kept at lOOmM stock solution in DMSO and was freshly dissolved in 1:100,000 carboxygenated artificial cerebrospinal fluid to the final concentration of lpM. For chronic injections Alzet osmotic pumps filled with marinobufagenin were used as described (Elkareh et al., 2007; Fedorova et al., 2009; Kennedy et al., 2006; Liu et al., 2012; Yoshika et al., 2007).

[075] Contextual fear conditioning: Mice were placed into a standard conditioning chamber and given a two-sec scrambled foot shock (0.75 mA) twice, two minutes and four minutes after session onset. The session ended 30 seconds after the second shock has been given. The occurrence of freezing was measured every 10 seconds throughout training. The mice were removed immediately and returned to their home cages. To test fear conditioning to the contextual cues, the mice were returned to the training context 24 hours after training for a five-minute test session. No shocks were presented during the test session. The occurrence of freezing was measured every 10 sec throughout testing.

[076] Morris water maze: MWM experiments were performed as previously described (Kaphzan et al., 2013). The paradigm consisted of 6 days of training (reference phase), where the mice were trained to locate a submerged hidden platform. Mice were given four trials per day (each trial for 60 s maximum, inter-trial interval 60 min). On the 7th day, a single-probe trial (probe test) was given after removing the hidden platform from the pool. The day after (8th day), mice were tested using a similar paradigm to find the platform by using a visible cue (visible test). The escape platform marked by a visible cue was moved randomly between four locations. The animals’ trajectories, escape latencies, number of previous platform position crossing, time spent in each quadrant, and swim speed were recorded with a computerized video-tracking system (Noldus EthoVision).

[077] Real-time Calcium imaging: Calcium dynamics of CA1 pyramidal cells from WT and AS mice was assessed by a multiphoton imaging system with quasi- ratiometric calcium imaging. Following a single stimulus of Schaffer collaterals synaptic activation, the calcium concentrations dynamics were measured in a reactive spine of a CA1 pyramidal cell. Slices were placed in perfusion chamber on the stage of an upright microscope (Olympus BX-61 WI) and were held in place with a non reactive plastic grid to avoid slice movement or field shift. To ensure prolonged viability of slices in imaging setup, slices were perfused with artificial CSF continuously bubbled with carboxygen gas mixture throughout the experiments. Fluo- 5F and Alexa Fluor 594 were filled in a patch pipette and applied to the cells by whole cell configuration. Excitation was performed at 8l0nm for two dyes Fluo-5F (green - calcium sensitive) and Alexa Fluor 594 (red - calcium insensitive) and the fluorescence dynamics was measured with a 500Hz line scan of Fluo-5F green (520nm) emission relative to the Alexa Fluor 594 emission (620nm). Data was acquired and analyzed by using Olympus fluoview FV1200.

EXAMPLE1

HFS during MBG 0.5uM application induces LTP deficit recovery in AS mice

[078] Application of 0.5mM MBG induced a recovery of LTP deficit in the AS mice (Fig. 1). Hippocampal slices from WT and AS mice were stimulated with a single train of high frequency stimulation (lsec lOOHz- indicated by an arrow). Some of the AS slices were treated with either vehicle or MBG 0.5mM 20 min before, during and 20 min after tetanization. WT vehicle: mice n=3, slices n=6; AS vehicle: mice n=2, slices n=4; AS MBG: mice n=2, slices n=4. P<0.05 for interaction of time and treatment in 2-way RM-ANOVA. Sample traces l-baseline, 2-20 min after HFS. MBG did not affect baseline synaptic transmission. Inhibition of al-NaKA with 0.5mM MBG is partial and mild.

EXAMPLE 2

Effect of MBG on resting membrane potential of CAl pyramidal cells

[079] The effect of al- NaKA inhibition with MBG IOmM on the resting membrane potential was tested (Fig. 2). Hippocampal slices were incubated for 10 min with MBG IOmM. Whole cell recordings of CA1 pyramidal cells show a larger effect on the resting membrane potential of AS mouse compared to a WT littermate. WT 1 mouse, cells n=4. AS: 1 mouse cells n=5. P=0.07 for the interaction in 2 way RM-ANOVA. MBG induced a larger effect in AS mice than in wild-type littermates, thus supporting the assumption of an increased al-NaKA activity

EXAMPLE 3

Chronic inhibition of al-NaKA activity rescues hippocampal dependent memory in the AS mice

[080] Osmotic pumps that chronically deliver lOpg/kg/day MBG by intraperitoneal (IP) infusion are implanted to mice. The MBG IP infusion starts 2 weeks before the initial contextual fear conditioning, and the memory testing is performed 24 hours after conditioning. Chronic IP infusion of MBG can correct the contextual fear memory deficit in AS model mice, 24 hours after training as described in the methods section. The infusions and experiments are performed in wild-type and AS mice littermates and MBG injected is compared to vehicle injection.

[081] IP infusion of MBG lOpg/kg/day corrects impaired contextual fear memory displayed by AS model mice. Such results are consistent with the idea that increased al- NaKA activity causes hippocampal dependent memory deficits in the AS model mice. Next, an additional set of experiments is performed by employing Morris water maze paradigm, to demonstrate that this intervention can also rescue hippocampal dependent visuospatial memory. The AS model mice exhibit impaired visuospatial memory in the Morris water maze and this impairment is corrected by MBG.

EXAMPLE 4

Chronic inhibition of al-NaKA activity rescues HFS-LTP in the AS mice

[082] Selective inhibition of al-NaKA corrects the HFS-LTP impairment in AS mice hippocampus. Hippocampal slices from AS model mice and their wild-type littermates are incubated with MBG ImM and high frequency stimulation LTP is induced as described in the Methods section. While there is no effect on baseline synaptic transmission, there is a rescue of the impaired LTP phenotype of the AS hippocampal slices. This is consistent with the genetic al-NaKA attenuation rescue.