EP1802314A2 - Compositions containing uridine, and methods utilizing same - Google Patents

Compositions containing uridine, and methods utilizing same

Info

Publication number
EP1802314A2
EP1802314A2 EP05796529A EP05796529A EP1802314A2 EP 1802314 A2 EP1802314 A2 EP 1802314A2 EP 05796529 A EP05796529 A EP 05796529A EP 05796529 A EP05796529 A EP 05796529A EP 1802314 A2 EP1802314 A2 EP 1802314A2
Authority
EP
European Patent Office
Prior art keywords
uridine
subject
choline
another embodiment
cell
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.)
Ceased
Application number
EP05796529A
Other languages
German (de)
French (fr)
Other versions
EP1802314A4 (en
Inventor
Richard J. Wurtman
Carol Watkins
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.)
Massachusetts Institute of Technology
Original Assignee
Massachusetts Institute of Technology
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
Priority claimed from US10/941,025 external-priority patent/US20050203053A1/en
Priority claimed from US10/944,269 external-priority patent/US8143234B2/en
Priority claimed from US10/972,777 external-priority patent/US8314064B2/en
Application filed by Massachusetts Institute of Technology filed Critical Massachusetts Institute of Technology
Publication of EP1802314A2 publication Critical patent/EP1802314A2/en
Publication of EP1802314A4 publication Critical patent/EP1802314A4/en
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • 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
    • A61K31/7068Compounds 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 having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds 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 having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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

Definitions

  • the present invention is directed to methods of improving cognitive and neurological functions and increasing synthesis and release of neurotransmitters and membrane synthesis by neural cells and brain cells, comprising administering a composition comprising a uridine or a source thereof.
  • Uridine is a pyrimidine nucleoside and is essential in the synthesis of ribonucleic acids and tissue glycogens such as UDP glucose and UTP glucose.
  • Prior medical uses of uridine alone include treatment of genetic disorders related to deficiencies of pyrimidine synthesis such as orotic aciduria.
  • Choline a dietary component of many foods, is part of several major phospholipids that are critical for normal membrane structure and function. Choline is included with lipid emulsions that deliver extra calories and essential fatty acids to patients receiving nutrition parenterally.
  • the present invention is directed to methods of improving cognitive and neurological functions and increasing synthesis and release of neuiotransmitters and membrane synthesis by neural cells and brain cells, comprising administering a composition comprising a uridine or a source thereof.
  • the present invention provides a method of improving a cognitive function in a subject, comprising administering to the subject a uridine, a source thereof, or a composition comprising a uridine and a choline.
  • the present invention provides a method of improving a neurological function in a subject, comprising administering to the subject a uridine, a source thereof, or a composition comprising a uridine and a choline.
  • the present invention provides a method of treating or ameliorating a decline in a cognitive function in a subject, comprising administering a uridine, a source thereof, or a composition comprising a uridine and a choline to the subject.
  • the present invention provides a method of increasing or enhancing an ability of a brain cell or a neural cell of a subject to synthesize a neurotransmitter, comprising administering to the subject or the brain cell or neural cell a uridine, a source thereof, or a composition comprising a uridine and a choline
  • the present invention provides a method of increasing a level of a neurotransmitter in a synapse, comprising contacting a neural cell adjacent to the synapse with a uridine, a source thereof, or a composition comprising a uridine and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a level of a neurotransmitter in a synapse.
  • the present invention provides a method of increasing a level of a cytidine in a tissue, plasma, or cell of a subject, comprising administering a uridine, a source thereof, to the subject.
  • the present invention provides a method of increasing a level of a cytidine in a tissue, plasma, or cell of a subject, comprising administering a composition comprising a uridine or a source thereof and a choline to the subject.
  • the present invention provides a method of stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject, comprising contacting the subject with a uridine, a source thereof, or a composition comprising a uridine and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject.
  • the present invention provides a method of stimulating or enhancing an outgrowth of a neurite of a neural cell, comprising contacting the neural cell with a uridine, a source thereof, or a composition comprising a uridine and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing an outgrowth of a neurite of a neural cell.
  • Figure 1 illustrates the coincidence of cytidine and tyrosine peaks (6.59) when tested by a standard HPLC method.
  • Figure 2 illustrates distinct cytidine (3.25) and tyrosine (2.92) peaks when tested by a modified HPLC method, which utilizes elution buffer with low methanol.
  • Figure 3 Oral UMP administration raises blood uridine levels in humans. Depicted is the ratio of uridine (set as 100% value) to cytidine in plasma after oral administration of 250 milligram per kg of body weight (mg/kg) of uridine.
  • Figure 4 Oral uridine administration raises blood uridine levels in gerbils. Depicted are plasma uridine levels 60 minutes following mock administration or administration of cytidine or uridine. **: p ⁇ 0.01 vs. mock-fed control; ##: p ⁇ 0.01 vs cytidine.
  • FIG. 5 Figure 5. Oral UMP administration raises blood uridine levels in gerbils. Depicted are plasma uridine levels at various time points following administration or administration of water or UMP.
  • FIG. 6 A UMP-supplemented diet raises blood uridine levels in gerbils. Depicted are plasma uridine levels in gerbils fed a diet containing the indicated percentages of UMP.
  • Figure 7 Oral uridine administration raises brain uridine levels. Depicted are brain uridine levels 60 minutes following mock administration or administration of cytidine or uridine. **: p ⁇ 0.01 vs. mock-fed control; ##: p ⁇ 0.01 vs cytidine.
  • FIG. 8 Figure 8. Oral UMP administration raises brain uridine levels Depicted are brain uridine levels at various time points following administration or administration of water or UMP.
  • Uridine is readily converted to cytidine in the brain. Depicted is the ratio of uridine (100%) to cytidine in plasma (A) and in the brain (B) after oral administration of 250 milligram per kg of body weight (mg/kg) of uridine
  • FIG. 10 Figure 10. Oral UMP administration raises brain CDP-choline levels. Depicted are brain CDP-choline levels at various time points following administration or administration of water or UMP.
  • FIG. 1 Uridine increases intracellular levels of CDP-choline in a neural cell line. Cells were incubated for 6 h with the indicated concentrations of uridine. Depicted are the means +/- S.E.M. of six dishes, expressed as picomole (pmol) CDP-choline/mg protein. The experiment was repeated 3 times. *: p ⁇ 0 05 [0024] Figure 12. UMP dietary supplementation significantly increases potassium-evoked dopamine (DA) release in striatal dialysate. (A) Effect of dietary UMP supplementations on K + - evoked striatal DA release.
  • Figure 14 Increased acetylcholine basal concentration with UMP treatment. Depicted are means +/- SEM, "*" denotes p value of >0.05.
  • Figure 15 Effect of UMP dietary supplemention on neurofilament protein levels in contralateral striatum.
  • A NF-70.
  • B NF-M *: p ⁇ 0,05, **: p ⁇ 0 01 compared to corresponding controls.
  • FIG. 16 Uridine treatment enhanced neurite outgrowth in PC 12 cells.
  • C Number of neuiites per cell after 2 or 4 days of NGF plus different concentrations of uridine (50, 100 and 200 ⁇ M).
  • E Levels of the structural proteins NF-70 and NF-M, as determined using Western blotting.
  • N NGF
  • U Uridine. Values represent means + SEM. **: p ⁇ 0 01, *** : p ⁇ 0.001 vs. NGF treatment.
  • Uridine treatment increased intracellular levels of UTP and CTP in PC 12 cells exposed to NGF for 2 days.
  • Uridine treatment 50 ⁇ M significantly increased intracellular UTP levels (A) and intracellular CTP levels (B) N - NGF,
  • U Uridine.
  • C Cytidine Values represent means + SEM * : p ⁇ 0.05 vs. NGF treatment
  • NGF-differentiated PC 12 cells express pyrimidine-sensitive P2Y receptors
  • FIG. 20 P2Y2 receptor co-localizes with the neuronal marker MAP-2.
  • Left panel P2Y2 receptor.
  • Middle panel MAP-2.
  • Right Panel Merge.
  • FIG. 21 P2Y receptor antagonists inhibited the effect of uridine on neurite outgrowth.
  • Cells were treated foi 4 days with NGF and with or without uridine (100 ⁇ M) and the P2Y receptoi antagonists PPADS, suramin, or RB-2. Values represent means + SEM. ***p ⁇ 0.001 vs. NGF treatment; Up ⁇ 0.05, ###p ⁇ 0 001 vs NGF plus uridine treatment.
  • FIG. 22 Phosphatidylinositol (Pl) turnover is stimulated by UTP and uridine.
  • Cells were metabolically labeled with [ 3 H] inositol overnight, stimulated with UTP, uridine, or UTP plus PPADS in the presence of lithium at the indicated concentrations, and radio-labeled inositol phosphates derived from PI breakdown were measured by scintillation counting. Values represent means + SEM. *p ⁇ 0-05, **p ⁇ 0,01 vs. control; #p ⁇ 0.05 vs. 100 ⁇ M UTP treatment.
  • Figure 23 Oral UMP improves learning and spatial memory in rats. 18-month old rats in restricted environments consumed a control diet or a UMP diet for 6 weeks, and then were tested, using a Morris Water Maze, 4 trials/day for 4 days. Mean time to locate the platform is given in seconds.
  • Figure 24 Oral UMP improves learning and spatial memory in gerbils Learning and spatial memory of gerbils fed a control diet or diets containing the indicated amount of UMP were tested in a radial arm maze. Results are depicted as the amount of time remaining before the 3-minute deadline.
  • Figure 25 Oral UMP improves working memory and reference memory.
  • the memory of gerbils fed a control or a 0.1% UMP diet for four weeks was tested using modification of the test depicted in Figure 24, which measured both working memory errors (A) and reference memory errors (B) Diamonds represent data points from control gerbils; triangles represent data points from gerbils fed 0.1% UMP diet [0038]
  • Figure 26 Uridine and choline increase neurotransmitter release in striatal slices (top panel), hippocampal slices (middle panel), and cortical slices (top panel). Data are expressed as nanomoles per milligram protein per two hour, and depicted as means ⁇ SEM.
  • the present invention is directed to methods of improving cognitive and neurological functions and increasing synthesis and release of neurotransmitters and membrane synthesis by neural cells and brain cells, comprising administering a composition comprising a uridine or a source thereof.
  • the present invention provides a method of improving a cognitive function in a subject, comprising administering to the subject a uridine or a source thereof, thereby improving a cognitive function in a subject
  • the present invention provides a method of improving a cognitive function in a subject, comprising administering to the subject a composition comprising a uridine or a source thereof and a choline, thereby improving a cognitive function in a subject.
  • uridine or a source thereof and a choline refers to 2 embodiments of the present invention: a) a combination of uridine and choline; b) a combination of a uridine source and choline.
  • uridine a combination of uridine and choline
  • uridine source a combination of a uridine source and choline.
  • the cognitive function is memory.
  • the memory is, in other embodiments, spatial memory, working memory, reference memory, short-term memory, long- term memory, or medium-term memory.
  • a ⁇ 6tHeFembo dime ⁇ f, ' the mem ⁇ ry i ⁇ fany dtHeFtype " of memory known in the art.
  • Each type of memory represents a separate embodiment of the present invention.
  • the data in Figures 21-23 show directly that uridine improves several types of memory.
  • the consistency of the effect across different species in different types of assessments of memory verifies the findings of the present invention
  • the data in Example 15 further show that the effects of uridine are enhanced by inclusion of a choline.
  • administration of compositions comprising uridine and choline are effective at improving memory- more effective, in one embodiment, than administration of either uridine or choline alone.
  • the cognitive function is learning
  • the learning is, in other embodiments, cognitive learning, affective learning, or psychomotor learning.
  • the learning is any other type of learning known in the art. Each type of learning represents a separate embodiment of the present invention.
  • the cognitive function is intelligence
  • the intelligence is linguistic intelligence, musical intelligence, spatial intelligence, bodily intelligence, interpersonal intelligence, intrapersonal intelligence, interpersonal intelligence, or logico-mathematical intelligence.
  • the intelligence is any other type of intelligence known in the art Each type of intelligence represents a separate embodiment of the present invention
  • the cognitive function is mental fitness. In another embodiment, the cognitive function is any other type of cognitive function known in the art. Each type of cognitive function represents a separate embodiment of the present invention.
  • "improving' " a cognitive function, or “improvement” of a cognitive function refer to increasing the capacity of the subject to perform the cognitive function.
  • the terms refer to an increased or improved baseline level of the cognitive fu ⁇ ction in the subject.
  • the terms refer to an increased or improved level of the cognitive function in response to a challenge or test.
  • improving a cognitive function refers to effecting a 10% improvement thereof In another embodiment, a 20% improvement is attained In other embodiments, a 30% improvement, a 40% improvement, a 50% improvement, a 60% improvement, a 70% improvement, an 80% improvement, or a 90% improvement is attained. In another embodiment, improving a cognitive function refers to effecting a 100% improvement thereof Each possibility represents a separate embodiment of the present invention.
  • improvement of a cognitive function is assessed relative to the cognitive function before beginning treatment. In anothei embodiment, improvement of a cognitive function is assessed relative to an untreated subject. In another embodiment, improvement of a cognitive function is assessed according to a standardized criterion such as, for example, a test or the like. Each type of improvement of cognitive activity represents a separate embodiment of the present invention
  • improvement of a cognitive function is assessed by the number of connections between neurons in the subject's brain.
  • the improvement is assessed by the number of capillaries in the subject ' s brain, or in a specific region of the subject's brain.
  • the improvement is assessed by neural activity.
  • the improvement is assessed by neural function, linguistic function, or ability to communicate
  • the improvement is assessed by measurement of levels of acetylcholine or other neurotransmitters or brain chemicals co ⁇ elated with cognitive function.
  • the improvement is assessed by Positron Emission Tomography (PET) scanning of the subject's brain, magnetic resonance imaging (MR! scanning of the subject's brain.
  • PET Positron Emission Tomography
  • MR magnetic resonance imaging
  • the improvement is assessed by Cognitive Abilities Screening Instrument (CASI) (Peila R et al, Stroke. 32: 2882-9, 2001).
  • CASI Cognitive Abilities Screening Instrument
  • the improvement is assessed by a test such as, for example, the tests disclosed herein (Example 13). Additional methods for assessing improvement of cognitive function are well known in the art, and are described, for example in Antonova E et al (Schizophr Res. 2004 Oct l ;70(2-3):117-45) and in Cognitive Function Analysis (Greenwood Pub Group, 1998). Each method represents a separate embodiment of the present invention
  • a composition of the present invention increases a level of cytidine, in the subject, thereby mediating one of the effects described herein (e.g improving cognitive or neurological function, stimulating neural function, membrane synthesis, neurotransmitter release, etc).
  • the effect is mediated by increasing a level of cytidine triphosphate (CTP) in the subject.
  • CTP cytidine triphosphate
  • the effect is mediated -by increasing a level of CDP-choline in the subject
  • the effect is mediated by increasing a level of a derivative of cytidine, CTP, CDP-choline in the subject.
  • the effect is mediated by increasing a level of a metabolite of cytidine, CTP, CDP-choline in the subject. In another embodiment, the effect is mediated without increasing a level of cytidine, CTP, CDP-choline, or a derivative or metabolite thereof.
  • Figures 9-1 1 show that orally administered uridine acts rapidly and effectively to raise levels of cytidine in the brain.
  • Figures 3-8 which show that uridine is effectively and rapidly absorbed into the bloodstream, in several species, including humans, these findings demonstrate that administration of uridine raises levels of cytidine, CTP, and CDP-choline.
  • the data in Example 15 further show that the effects of uridine are enhanced by inclusion of a choline.
  • the cytidine level is a systemic level. In another embodiment, the cytidine level is a brain level In another embodiment, the cytidine level is a nervous system level. Each possibility represents a separate embodiment of the present invention.
  • the potential benefit of uridine administration is greater than the benefit of cytidine administration This is due to the fact that cytidine, as opposed to uridine, either cannot cross or is much less efficient than uridine in crossing the blood-brain barrier (Cornford et al., Independent blood-brain barrier transport systems for nucleic acid precursors. Biochim. Biophys. Acta 349:21 1-219, 1975).
  • the increase in cytidine, CTP, or CDP-choline or a derivative or metabolite thereof enables the cell to increase levels of a phospholipid, thereby mediating one of the effects described herein.
  • the phospholipid is phosphatidylcholine (PC)
  • the phospholipid is phosphatidylethanolamine (PE).
  • the phospholipid is phosphatidylserine (PS).
  • the phospholipid is or a derivative or metabolite of PC, PE, oi PS Each possibility represents a separate embodiment of the present invention.
  • the present invention provides a method of improving a neurological function in a subject, comprising administering to the subject a uridine or a source thereof, thereby improving a neurological function in a subject.
  • the present invention provides a method of improving a neurological function in a subject, comprising administering to the subject a composition comprising a uridine or a source thereof and a choline, thereby improving a neurological function in a subject.
  • the neurological function that is improved by a method of the present invention is a synaptic transmission.
  • the synaptic transmission is adjacent to a motor neuron.
  • the synaptic transmission is adjacent to an intemeuron.
  • the synaptic transmission is adjacent to a sensory neuron
  • Each type of synaptic transmission represents a separate embodiment of the present invention.
  • the synaptic transmission is improved or enhanced by means of stimulating oi enhancing an outgrowth of a neurite of a neural cell.
  • stimulating or enhancing an outgrowth of a neurite of a neural cell is partially responsible for improving or enhancing the synaptic transmission.
  • a composition of the present invention improves or enhances synaptic transmission without stimulating an outgrowth of a neurite.
  • Neuron refers, in one embodiment, to a process growing out of a neuron In one embodiment, the process is a dendrite. In another embodiment, the process is an axon Each type of neurite represents a separate embodiment of the present invention.
  • the synaptic transmission is improved or enhanced by increasing the number of neurites of the neural cell.
  • improvement or enhancement of the synaptic transmission occurs without increasing the number of neurites of the neural cell.
  • the synaptic transmission is improved oi enhanced by stimulating or enhancing blanching of a neurite of a neural cell
  • improvement or enhancement of the synaptic transmission occurs without stimulating or enhancing branching of a neurite of a neural cell
  • Example 9 The data of Example 9 shows that when levels of membrane precursors are increased, neurons produce moie neurites, with more branches. By increasing its surface aiea and size, a cell is able, in one embodiment, to form more connections with neighboring cells. Moreover, an increase in the amount or composition of plasma membiane alters, in one embodiment, neurotransmitter synthesis and release, which also, in one embodiment, affects memory formation. Thus, compounds that promote neurite outgrowth, such as uridine, are useful for treatment of neurodegenerative disorders like Alzheimer's disease, which involves loss of neuronal connections and memory impairment.
  • improving the synaptic transmission in the subject is achieved by increasing an amount of a membrane of a neural cell as a result of administration of the uridine and/or choline.
  • the improvement is achieved by stimulating a synthesis of a membrane of a neural cell.
  • the improvement is achieved by enhancing a synthesis of a membrane of a neural cell.
  • stimulating or enhancing an amount of or a synthesis of a membrane of a neural cell is partially responsible for mediating improving the synaptic transmission in the subject.
  • the uridine and/or choline improves the synaptic transmission without stimulating or enhancing an amount of or a synthesis of a membrane of a neural cell.
  • the neurological function that is improved or enhanced is a function of a neurotransmitter.
  • the improvement occurs by means of increasing a level of the neurotransmitter in a synapse.
  • the improvement occurs by means of increasing the release of the neurotransmitter into a synapse.
  • the improvement occurs without changing the level or release of the neurotransmitter in a synapse.
  • the data in Figures 12-13 show that uridine significantly improves neurotransmitter function, highlighting the ability of uridine to improve neurological function.
  • the data in Figures 14-17 show a beneficial effect of uridine on the morphology of neurites, further demonstrating the ability of uridine to improve neurological function.
  • the data in Example 15 further show that the effects of uridine are enhanced by inclusion of a choline
  • administration of compositions comprising uridine and choline are effective at improving neurological function - more effective, in one embodiment, than administration of either uridine or choline alone.
  • the present invention provides a method of treating or ameliorating a decline in a cognitive function in a subject, comprising administering a uridine or a source thereof to the subject, thereby treating or ameliorating a decline in a cognitive function in a subject.
  • the present invention provides a method of treating or ameliorating a decline in a cognitive function in a subject, comprising administering a composition comprising a uridine or a source thereof and a choline to the subject, thereby inhibiting or preventing a decline in a cognitive function in a subject
  • Treating or ameliorating a decline in a cognitive function refers, in one embodiment, to mitigating the decline
  • the phrase refers to preventing the decline.
  • the phrase refers to ieversing the decline.
  • the phrase refers to slowing the decline.
  • the phrase refers to halting the decline
  • the decline in a cognitive function results from a neurological disorder.
  • the neurological disorder is a memory disorder
  • the memory disorder comprises, in one embodiment, a memory decline.
  • the memory decline is associated with brain aging.
  • the memory disorder is selected from Pick's disease, Lewy Body disease, or a dementia.
  • the dementia is associated with Huntington's disease or AIDS dementia.
  • Each possibility represents a separate embodiment of the present invention.
  • the decline in a cognitive function results from a neurodegenerative disease.
  • the neurodegenerative disease is Alzheimer's disease.
  • the neurodegenerative disease is amyotrophic lateral sclerosis, multiple system atrophy, Parkinson's disease, progressive supranuclear palsy, frontotemporal dementia, Huntington's disease, or a prion disease.
  • the neurodegenerative disease is any other neurodegenerative disease known in the art. Eacli possibility represents a separate embodiment of the present invention
  • the decline in a cognitive function results from a cardiovascular disease
  • the cardiovascular disease is a stroke
  • the cardiovascular disease is a multi-infarct dementia.
  • the cardiovascular disease is any other cardiovascular disease known in the art. Each possibility represents a separate embodiment of the present invention.
  • the neurological disorder is associated with a dopaminergic pathway In another embodiment, the neurological disorder is not associated with a dopaminergic pathway Each possibility represents a separate embodiment of the present invention.
  • the neurological disorder is a cognitive dysfunction. In one embodiment, the cognitive dysfunction is dyslexia. In other embodiments, the cognitive dysfunction comprises a lack of attention, a lack of alertness, a lack of concentration, or a lack of focus. In other embodiments, the cognitive dysfunction comprises minimal cognitive impairment or age-ielated memory impairment. Each possibility represents a separate embodiment of the present invention.
  • the neurological disorder is an emotional disorder.
  • the emotional disorder comprises mania, depression, stress, panic, anxiety, dysthymia, or psychosis.
  • the emotional disorder comprises a seasonal effective disorder.
  • the emotional disorder comprises a bipolar disorder.
  • the neurological disorder is a psychiatric disease.
  • the neurological disorder is a depression.
  • the depression is an endogenous depression.
  • the depression is a major depressive disorder.
  • the depression is depression with anxiety.
  • the depression is bipolar depression. Each type of depression iepiesents a separate embodiment of the present invention.
  • the neurological disorder is selected from the group consisting of ataxia and Friedreich's ataxia
  • the neurological disorder is a movement disorder.
  • the movement disorder comprises, in other embodiments, a tardive dyskinesia, a dystonia, or a Tourette's syndrome.
  • the movement disorder is any other movement disorder known in the art
  • the neurological disorder is a cerebrovascular disease.
  • the cerebro-vascular disease results, in one embodiment, from hypoxia In another embodiment, the disease results from any other cause capable of causing a cerebro-vascular disease.
  • the disease is cerebral thrombosis. In another embodiment, the cerebro-vascular disease is ischemia.
  • the neurological disorder is a behavioral syndrome.
  • the neurological disorder is a neurological syndrome.
  • the behavioral syndrome or neurological syndrome follows brain trauma, spinal cord injury, or anoxia.
  • the neurological disorder is a peripheral nervous system disorder.
  • the peripheral nervous system disorder is a neuromuscular disorder, myasthenia gravis, or post-polio syndrome.
  • the peripheral nervous system disorder is any other peripheral nervous system disorder known in the art.
  • the neuromuscular disorder is a muscular dystrophy
  • the present invention provides a method of increasing or enhancing an ability of a brain cell or a neuial cell of a subject to synthesize a neurotransmitter, comprising administering to the subject or the brain cell or neural cell a uridine or a source thereof, thereby increasing or enhancing an ability of a brain cell of a subject to synthesize a neurotransmitter.
  • the present invention provides a method of increasing or enhancing an ability of a brain cell or a neural cell of a subject to synthesize a neurotransmitter, comprising administering to the subject or the brain cell or neural cell a composition comprising a uridine or a source thereof and a choline, thereby increasing or enhancing an ability of a brain cell of a subject to synthesize a neurotransmitter.
  • the present invention provides a method of increasing or enhancing an ability of a brain cell or a neural cell of a subject to repeatedly release an effective quantity of a neurotransmitter into a synapse, comprising administering to the subject or the brain cell or neural cell with a uridine or a source thereof, thereby increasing or enhancing an ability of a brain cell or a neural cell of a subject to repeatedly release an effective quantity of a neurotransmitter into a synapse.
  • th “ e” present “ invention provides a " method of increasing “ of " enhancing an ability of a brain eel) or a neural cell of a subject to repeatedly release an effective quantity of a neurotransmitter into a synapse, comprising administering to the subject or the brain cell or neural cell with a composition comprising a uridine or a source thereof and a choline, thereby increasing or enhancing an ability of a brain cell or a neural cell of a subject to repeatedly release an effective quantity of a neurotransmitter into a synapse,
  • findings of the present invention show that uridine enhances the ability of neurons to synthesize neurotransmitters and repeatedly release them (Example 7)
  • the data in Example 15 further show that this effect of uridine is enhanced by inclusion of choline.
  • the release which is enhanced by a method of the present invention occurs following a stimulation of the neuron. In one embodiment, the release which is enhanced occurs following a depolarization of the neuron. In one embodiment, the release which is enhanced is a basal neurotransmitter release. In one embodiment, the stimulation of the neuron comprises exposure of the neuron to a potassium ion. In another embodiment, the stimulation of the neuron comprises any other means of neural stimulation known in the art. Methods for assessing neural stimulation and release of neurotransmitters are well known in the art, and are described, for example, in Bewick GS, J Neurocytol 32: 473-87, 2003. Each possibility represents a separate embodiment of the present invention
  • the picsent invention provides a method of increasing a level of a neurotransmitter in a synapse, comprising contacting a neural cell adjacent to the synapse with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a level of a neurotransmitter in a synapse.
  • the present invention provides a method of increasing a level of a neurotransmitter in a synapse, comprising contacting a neural cell adjacent to the synapse with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a level of a neurotransmitter in a synapse.
  • the present invention provides a method of increasing a sensitivity of a neuron to a stimulus, comprising contacting the neuron with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a sensitivity of a neuron to a stimulus,
  • the present invention provides a method of increasing a sensitivity of a neuron to a stimulus, comprising contacting the neuron with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a sensitivity of a neuron to a stimulus.
  • the neurotransmitter whose levels or activity, or release is affected by methods of the present invention is acetylcholine.
  • the neurotransmitter is dopamine
  • the neurotransmitter is serotonin.
  • the neurotransmitter is 5-hydroxytryptamine (5-HT).
  • the neurotransmitter is GABA.
  • the neurotransmitter is any other neurotransmitter known in the art.
  • Each type of neurotransmitter represents a separate embodiment of the present invention
  • the present invention provides a method of stimulating a production of a phosphatidylcholine (PC) by a brain cell or neural cell of a subject, compiising administering to the subject or brain cell or neural cell a uridine or a source thereof, thereby stimulating a production of a PC by a brain cell or neural cell.
  • PC phosphatidylcholine
  • the present invention provides a method of stimulating a production of a phosphatidylcholine (PC) by a brain cell or neural cell of a subject, comprising administering to the subject or brain cell or neural cell a composition comprising a uridine or a source thereof and a choline, thereby stimulating a production of a PC by a brain cell or neural cell.
  • a composition comprising a uridine or a source thereof and a choline, thereby stimulating a production of a PC by a brain cell or neural cell.
  • findings of the present invention show that uridine enhances synthesis of the PC precursor CDP-choline (Example 6).
  • the data in Example 15 further show that this effect of uridine is enhanced by inclusion of choline.
  • the present invention provides a method of stimulating or enhancing an amount of or a synthesis of a component of a cell membrane, comprising contacting the cell with a uridine or a source theieof, thereby stimulating or enhancing an amount of or a synthesis of a cell membrane.
  • the present invention provides a method of stimulating or enhancing an amount of or a synthesis of a component of a cell membrane, comprising contacting the cell with a composition comprising a uridine or a source thereof and a choline, thereby stimulating or enhancing an amount of or a synthesis of a cell membrane.
  • the component whose synthesis is enhanced by a method of the present invention is a PC.
  • the component is a glyceiophospholipid.
  • the component is a phosphatide acid
  • the component is a PE
  • the component is a lecithin.
  • the component is a Pl.
  • the component is a PS
  • the component is a 2-lysolecithin, a plasmalogen, a choline plasmalogen, a phosphatidylglycerol, a choline diphosphatidylglycerol, a choline sphingolipid, or a choline sphingomyelin.
  • the component is any other phospholipid known in the art Each type of phospholipid represents a separate embodiment of the present invention.
  • the present invention provides a method of stimulating or enhancing an amount of or a synthesis of a phospholipid precursor, comprising contacting the cell with a uridine or a source thereof, thereby stimulating or enhancing an amount of or a synthesis of a phospholipid precursor.
  • the present invention provides a method of stimulating or enhancing an amount of or a synthesis of a phospholipid precursor, comprising contacting the cell with a composition comprising a uridine or a source thereof and a choline, thereby stimulating or enhancing an amount of or a synthesis of a phospholipid precursor.
  • the phospholipid precursor is CDP-choline (Example 6).
  • the phospholipid precursor is CTP
  • the phospholipid precursor is inositol.
  • the phospholipid precursor is choline
  • the phospholipid precursor is glycerol.
  • the phospholipid precursor is acetate
  • the phospholipid precursor is any other phospholipid precursor known in the art. Each phospholipid precursor represents a separate embodiment of the present invention.
  • the present invention provides a method of stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject, comprising contacting the subject with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject
  • the present invention provides a method of stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject, coi ⁇ prising " contacting the subject with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject
  • the membrane is a neurite membrane.
  • the membrane is a dendritic membrane.
  • the membrane is an axonal membrane.
  • the membrane is any other type of membiane known in the art. Each type of membrane represents a separate embodiment of the present invention.
  • stimulating an amount of or a synthesis of the cell membrane is accomplished by stimulating or enhancing a synthesis of a phospholipid (Example 6).
  • stimulating or enhancing an amount of or a synthesis of a membrane of a neural cell is accomplished by stimulating or enhancing a synthesis of a phospholipid precursor (Example 6)
  • stimulating oi enhancing a synthesis of a phospholipid or a precursor thereof is partially responsible for stimulating an amount of or a synthesis of a membrane of a neural cell
  • a composition of the present invention stimulates the amount of or a synthesis of a membrane without stimulating or enhancing a synthesis of a phospholipid or a precursor thereof
  • membrane production is assessed by measuring the level of neurite outgrowth or branching (Example 9).
  • membrane production is assessed by measuring the level of a membrane maikei protein (Example 8).
  • membrane production is assessed by measuring synthesis of a membrane precursor.
  • membrane production is assessed by measuring amounts of membrane prior to and following uridine treatment.
  • membrane production is assessed by measuring biological indicators of membrane turnover.
  • Indicators or cellular membrane tumovei are well known in the art, and are described, for example, in Das KP et al, Neurotoxicol Teratol 26(3): 397-406, 2004 Each method of assessing membrane production represents a separate embodiment of the present invention.
  • the present invention provides a method of stimulating or enhancing an outgrowth of a neurite of a neural cell, comprising contacting the neural cell with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing an outgrowth of a neurite of a neural cell
  • the present invention provides a method of stimulating or enhancing an outgrowth of a neurite of a neural cell, comprising contacting the neural cell with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing an outgrowth of a neurite of a neural cell.
  • findings of the present invention show that uridine enhances outgrowth and branching of neurites (Example 9). The data in Example 15 further show that this effect of uridine is enhanced by inclusion of choline.
  • the present invention provides a method of increasing a number of neurites of a neuial cell, comprising contacting the neural cell with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a number of neurites of a neural cell.
  • the present invention provides a method of increasing a number of neurites of a neural cell, comprising contacting the neural cell with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a number of neurites of a neural cell
  • the present invention provides a method of stimulating or enhancing a branching of a neurite of a neural cell, comprising contacting the neural cell with a uridine or a source thereof, thereby stimulating or enhancing a branching of a neurite of a neural cell
  • the present invention provides a method of stimulating or enhancing a branching of a neurite of a neural cell, comprising contacting the neural cell with a composition comprising a uridine or a source thereof and/or a choline, thereby stimulating or enhancing a branching of a neurite of a neural cell
  • the cell that is the target of methods of the present invention or is contacted in the methods is a neural cell.
  • the cell is a brain cell
  • the cell is any cell in which synthesis of a membrane or a component thereof is enhanced by contact with a composition comprising a uridine and/or a choline.
  • the cell is any cell in which a neurological function is enhanced by contact with a composition comprising a uridine and/or a choline.
  • the neural cell, neuiite, or brain cell of methods of the present invention is newly differentiated.
  • the cell is not newly differentiated.
  • "newly differentiated " ' refers to a neuron that has differentiated in the 24 hours prior to commencing administration of the uridine and/or choline
  • the neuron has differentiated in the 48 hours prior to administration.
  • the neuron has differentiated in the 72 hours prior to administration
  • the neuron has differentiated in the 1 week prior to administration.
  • "newly differentiated " ' refers to a neuion that completes its differentiation following commencement of administration of the composition of the present invention. Each possibility represents a separate embodiment of the present invention.
  • the present invention provides a method of increasing a level of a cytidine in a tissue, plasma, or cell of a subject, comprising administering a uridine or a source thereof to the subject, thereby increasing a level of a cytidine in a tissue, plasma, or cell
  • the present invention provides a method of increasing a level of a cytidine in a tissue, plasma, or cell of a subject, comprising administering a composition comprising a uridine or a source thereof and a choline to the subject, thereby increasing a level of a cytidine in a tissue, plasma, or cell.
  • the piesent invention provides a method of increasing a level of a CTP in a tissue, plasma, or cell of a subject, comprising administering a composition of the present invention to the subject
  • the present invention provides a method of increasing a level of a CDP- cholme in a tissue, plasma; or ceil of a subject, compri sing-administering a composition of the present invention.
  • the present invention provides a method of increasing a level of a derivative of a cytidine, a CTP, or a CDP-choline in a tissue, plasma, or cell of a subject, comprising administering a composition of the present invention
  • the present invention provides a method of increasing a level of a metabolite of a cytidine, a CTP, or a CDP-choline in a tissue, plasma, or cell of a subject, comprising administering a composition of the present invention.
  • the tissue is a brain tissue. In one embodiment, the tissue is a neural tissue. In another embodiment, the tissue is a spinal tissue. In another embodiment, the tissue is any other tissue known in the art.
  • the cell is a brain cell. In one embodiment, the cell is a neural cell. In another embodiment, the cell is a spinal cell. In another embodiment, the cell is any other cell known in the art. Each possibility represents a separate embodiment of the present invention.
  • the uridine that is administered in the present invention is a uridine-5'-monophosphate (UMP).
  • the uridine is a uridine-5 " - diphosphate (UDP)
  • the uridine is a uridine-5 " -triphosphate (UTP).
  • the uridine is UDP glucose.
  • a uridine precursor is administered in methods of the present invention
  • the uridine precursor that is administered is a cytidine- 5'-monophosphate.
  • the uridine precursor that is administered is a cytidine-5' -diphosphate (CDP).
  • the uridine precursor that is administered is a CDP-glucose.
  • the uridine precursor that is administered is any pharmacologically acceptable uridine precursor, deiivative or metabolite known in the art.
  • a uridine derivative is administered in methods of the present invention.
  • the term "derivative" in one embodiment refers to a compound chemically related to uridine in such a way that uridine is converted to the derivative in a subject's body.
  • “derivative” refers to a compound chemically related to uridine in such a way that the derivative is converted to uridine in a subject's body
  • the conversion occurs via one or more stable intermediates.
  • the conversion occurs directly.
  • a uridine metabolite is administered in methods of the present invention
  • uridine-based compounds other than uridine itself serve as uridine sources or uridine precursors. These are, in some embodiments, uridine-rich food or dietary products like algae; salts of uridine like uridine phosphates, acylated uridine or the like.
  • therapeutically or pharmacologically effective doses of acyl derivatives of uridine or mixtures theieof, e.g. those disclosed in U.S. Pat. No. 5,470,838, are administered.
  • the uridine sourse is cytidine-diphosphocholine (CDP- choline; citicholine). While citicholine contains choline as well as uridine in a 1 :1 molar ratio, it is not, in one embodiment, sufficient to supply all the choline required by the subject. Thus, in this embodiment, citicholine serves a the source of all the uridine and some of the choline required by the subject.
  • a salt of the uridine piecursor, derivative or source is utilized in a method of the present invention
  • the salt is UMP disodium (Examples 2-3)
  • the salt is any other pharmacologically acceptable salt of a uridine precursor or derivative.
  • the composition that is administered comprises the salt of the uiidine or precursor or derivative thereof as the sole active ingredient.
  • Each uridine salt rcpiesents a separate embodiment of the present invention.
  • a mixture of two oi more of the above uridine-related compounds is administered.
  • Each type of uridine precursor, derivative, metabolite, or source represents a separate embodiment of the present invention.
  • uridine' refers, in one embodiment, to any uridine phosphate, uridine precursor, uridine metabolite, uridine-based compound, oi salt thereof mentioned above.
  • uridine refers to any uridine or related compound that is known in the art.
  • the uridine, derivative, source, or precursor thereof is administered in methods of the present invention in a dosage of between about 20 milligrams (mg) and 50 giams (g) per day.
  • the uridine or related compound is administered in a dosage of about 50 mg-30 g per day.
  • the dosage is about 75 mg-20 g; 100 mg-20 g; 100 mg-10 g; 200 mg-8 g; 400 mg-6 g; 600 mg-4 g; 800 mg-3 g; 1 -2.5 g; 1 5-2 g; 5 mg-5 g; or 5 mg-50 g per day.
  • Each dosage or dosage range represents a separate embodiment of the present invention.
  • the choline administered in methods of the present invention is a choline salt.
  • the salt is choline chloride.
  • the salt is choline bitartrate.
  • the salt is choline stearate.
  • the salt is choline alfoscerate, choline dehydrocholate., choline dihydrogen citrate, or choline salicylate
  • the salt is any other choline salt known in the art.
  • the choline is a choline-based compound, e.g. a choline ester.
  • the choline is a compound that dissociates to choline.
  • the compound is sphingomyelin
  • the compound is an acylglycerophosphocholine.
  • the compound is lecithin.
  • the compound is lysolecithin.
  • the compound is glycerophosphatidylcholine.
  • a mixture of two or more of the above choline-related compounds is administered
  • choline refers, in one embodiment, to any choline phosphate, choline precursor, choline metabolite, choline-based compound, or salt thereof mentioned above
  • choline refers to any choline or related compound that is known in the art
  • the choline or choline-related compound is administered in such a manner and dosage that a choline level of at least 20-30 nanomoles is attained in the subject's blood or brain In another embodiment, a choline level of 10-50 nanomoles is attained. In another embodiment, a choline level of 5-75 nanomoles is attained. In another embodiment, a choline level of 25-40 nanomoles is attained. In another embodiment, a choline level of 30-35 nanomoles is attained. Each possibility represents a separate embodiment of the present invention.
  • the choline, derivative, source, or precursor thereof is administered in methods of the present invention in a dosage of 20 mg-50 g per day
  • the choline or related compound is administered in a dosage of about 50 mg-30 g; 75 mg-20 g; 100 mg-20 g; 100 mg-10 g; 200 mg-8 g; 400 mg-6 g; 600 mg-4 g; 800 mg-3 g; 1 -2.5 g; 1 ,5-2 g; 5 mg-5 g; or 5 mg-50 g per day.
  • Each dosage range represents a separate embodiment of the present invention.
  • a composition of the present invention is administered at a dose that produces a desired effect in at least 10% of a population of treated patients.
  • the dose is that which produces the effect in at least 20% of treated patients.
  • the effect is produced in at least 30%, in at least 40%, in at least 50%, in at least 60%, in at least 70%, in at least 80%, or in at least 90% of the treated patients.
  • the effect is produced in over 90% of the patients.
  • the subject of methods of the present invention is a mammal.
  • the subject is a human.
  • the subject is a rodent or a laboratory animal.
  • the subject is a male.
  • the subject is a female.
  • the subject is any other type of subject known in the art. Each possibility represents a separate embodiment of the present invention.
  • administering refers to bringing a subject in contact with a compound of the present invention.
  • administration comprises swallowing or imbibing the composition of the present invention.
  • the step of administration utilizes a pharmaceutical composition, a nutritional supplement, or the like. Each possibility represents a separate embodiment of the present invention.
  • administration is performed by the subject. In another embodiment, administration is performed by a care provider. In another embodiment, administration is performed by a third party Each type of administration represents a separate embodiment of the present invention.
  • an additional therapeutic compound is administered to the subject as part of the method of the present invention.
  • the uridine or precursor, derivative or source thereof is the sole active ingredient in the composition.
  • the uridine or precursor, derivative or source thereof and choline or precursor, derivative or source thereof are the sole active ingredients in the composition.
  • the additional therapeutic compound is a drug that acts as a undine phosphorylase inhibitor; e.g. benzyl barbiturate or derivatives thereof.
  • the compound is a drug that increases uridine availability.
  • the compound is a uridine secretion-inhibiting compound, e.g.
  • the compound is a uridine renal transport competitors, e.g. L-uridine, L- 2',3'-dideoxyuridine, and D-2',3'-dideoxyuridine.
  • the compound is a drug which acts in synergy with uridine in generation of a phospholipid.
  • the compound is a compound which competes with uridine in kidney clearance, e.g. L-uridine, L-2',3'-dideoxyuridine, and D-2',3'-dideoxyuridine or mixtures thereof as disclosed in U.S. Pat. Nos. 5,723,449 and 5,567,689.
  • the compound is any other compound that is beneficial to a subject
  • a method of the present invention causes one of the above effects by means of stimulating a P2Y receptor of a neural cell, neuron, or brain cell.
  • one of the above effects is caused partially as a result of stimulating a P2Y receptor of a neural cell or neuron.
  • one of the above effects is caused partially or fully by means of stimulating a P2Y receptor of another cell type.
  • one of the above effects is caused without stimulating a P2Y receptor.
  • the stimulation of a P2Y receptor is mediated by uridine or a related compound in a composition of the present invention.
  • the uridine is converted to a second compound that stimulates a P2Y receptor in the cell.
  • the second compound is uridine-5'-triphosphate.
  • the second compound is any metabolic product known in the art of uridine or derivative or source thereof. Each compound represents a separate embodiment of the present invention.
  • the uridine or derivative or source thereof is converted into the second compound intracellularly or extracellularly
  • the uridine or derivative or source thereof is secreted from a cell after being converted into the second compound.
  • the uridine or derivative or source -thereof contacts a different cell after being- secreted from the cell in which it was converted to the second compound, and stimulates a P2Y receptor in the different cell.
  • P2Y receptors are a family of receptors known to be involved in platelet activation and other biological functions. They are reviewed in Mahaul-Smith MP et al, Platelets. 2004 15 :131 -44, 2004
  • the P2Y receptor of the present invention is a P2Y2 receptor.
  • the P2Y receptor is a P2Y4 receptor.
  • the P2Y receptor is a P2Y6 receptor.
  • the P2Y receptor is any other P2Y receptor known in the art. Each possibility represents a separate embodiment of the present invention.
  • the P2Y receptor stimulates a second messenger
  • the second messenger is a G alpha protein.
  • the second messenger is a G alpha(q) protein.
  • the second messenger is cAMP.
  • the second messenger is any other second messenger known in the art.
  • Second messengers, and their associated signaling pathways, are well known in the art, and are described, for example, in Ferguson S, Pharm Rev 53: 1 -24, 2001 ; Huang E et al, Ann Rev Biochem 72: 609-642, 2003; and Blitterswijk W et al, Biochem J. 369: 199-21 1 , 2003.
  • Each second messenger represents a separate embodiment of the present invention
  • the second messenger stimulates a phospholipase C enzyme, modulates intracellular calcium levels, or increases protein kinase C activity.
  • one or more of the above pathways stimulates membrane production.
  • the second messenger modulates or stimulates another cellular pathway that stimulates membrane production.
  • uridine or a related compound in a composition of the present invention stimulates a receptor other than a P2Y receptor
  • the uridine and/or choline is carried in the subjects' bloodstream to the subject's brain cell or neural cell.
  • the substance is carried by diffusion to the subject's brain cell or neural cell.
  • the substance is carried by active transport to the subject's brain cell or neural cell.
  • the substance is administered to the subject in such a way that it directly contacts the subject ' s brain cell or neural cell.
  • pharmaceutical composition refers to a therapeutically effective amount of the active ingredients, i.e the uridine and/or choline, together with a pharmaceutically acceptable carrier or diluent.
  • “Therapeutically effective amount” refers to that amount which provides a therapeutic effect for a given condition and administration regimen
  • the pharmaceutical composition containing the uridine and/or choline is administered to a subject by any method known to a person skilled in the art, such as parenterally, paracancerally, transmucosally, transdermal Iy, intramuscularly, intravenously, intradermally, subcutaneously, intraperitonealy, intraventricular ⁇ , intracranially, intravaginally or intratumoraUy.
  • the pharmaceutical compositions are administered oially, and thus is formulated in a form suitable for oral administration, i.e. as a solid or a liquid preparation.
  • suitable solid oral formulations include, for example, tablets, capsules, pills, granules, pellets and the like.
  • Suitable liquid oral formulations include solutions, suspensions, dispersions, emulsions, oils and the like.
  • the composition containing the uridine and choline is formulated in a capsule.
  • the compositions of the present invention comprises a hard gelating capsule, in addition to the active compounds and the inert carrier or diluent.
  • the pharmaceutical compositions are administered by intravenous, intraarterial, or intramuscular injection of a liquid preparation
  • suitable liquid formulations include solutions, suspensions, dispersions, emulsions, oils and the like.
  • the pharmaceutical compositions are administered intravenously, and are thus formulated in a form suitable for intravenous administration.
  • the pharmaceutical compositions are administered intraarterial Iy, and are thus formulated in a form suitable for intraarterial administration
  • the pharmaceutical compositions are administered intramuscularly, and are thus formulated in a form suitable for intramuscular administration.
  • the pharmaceutical compositions are administered as a suppository, for example a rectal suppository or a urethral suppository.
  • the pharmaceutical compositions are administered by subcutaneous implantation of a pellet.
  • the pellet provides for controlled release of uridine and/or choline over a period of time.
  • Pharmaceutically acceptable carriers or diluents are well known to those skilled in the art.
  • the carrier or diluent is, in one embodiment, a solid carrier or diluent for solid formulations, a liquid carrier or diluent for liquid formulations, or mixtures thereof.
  • Solid ca ⁇ iers/diluents include, in other embodiments, a gum, a starch (e.g. corn starch, pregeletanized starch), a sugar (e.g., lactose, mannitol, sucrose, dextrose), a cellulosic material (e.g. microcrystalline cellulose), an acrylate (e.g. polymethylacrylate), calcium carbonate, magnesium oxide, talc, or mixtures thereof.
  • a starch e.g. corn starch, pregeletanized starch
  • a sugar e.g., lactose, mannitol, sucrose, dextrose
  • a cellulosic material e.g. microcrystalline cellulose
  • an acrylate e.g. polymethylacrylate
  • pharmaceutically acceptable carriers are, in other embodiments, aqueous oi non-aqueous solutions, suspensions, emulsions or oils.
  • Non-aqueous solvents include propylene glycol, polyethylene glycol, and injectable organic esters such as ethyl oleate.
  • Aqueous earners include water, alcoholic/aqueous solutions, emulsions oi suspensions, including saline and buffered media
  • oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, mineral oil, olive oil, sunflower oil, and fish-liver oil.
  • compositions further comprise binders (e.g. acacia, cornstarch, gelatin, carbomer, ethyl cellulose, guar gum, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, povidone), disintegrating s (e g cornstarch, potato starch, alginic acid, silicon dioxide, croscarmelose sodium, crospovidone, guar gum, sodium starch glycolate), buffers (e.g., Tris-HCL, acetate, phosphate) of various pH and ionic strength, additives such as albumin or gelatin to prevent absorption to surfaces, detergents (e.g., Tween 20, Tween 80, PIuronic F68, bile acid salts), protease inhibitors, surfactants (e g sodium lauryl sulfate), permeation enhancers, solubilizers (e.g., glycerol, polyethylene glycerol), solubilizers (e
  • the pharmaceutical compositions provided herein are controlled release compositions, i.e. compositions in which the uridine and/or choline is released over a period of time after administration.
  • Controlled or sustained release compositions include formulation in lipophilic depots (e.g. fatty acids, waxes, oils).
  • the composition is an immediate release composition, i.e. a composition in which all of the uridine and/or choline is released immediately after administration
  • the pharmaceutical composition is delivered in a controlled release system.
  • the composition is administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump is used (see Langer, supra; Sefton, CRC Crit Ref Biomed Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J Med. 321 :574 (1989).
  • polymeric materials are used.
  • a controlled release system is placed in proximity to the theiapeutic target, i.e., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol 2, pp. 1 15-138 (1984) Other controlled release systems are discussed in the review by Langer (Science 249.1527-1533 (1990).
  • compositions which contain an active component are well understood in the art, foi example by mixing, granulating, or tablet-forming processes.
  • the active therapeutic ingredient is often mixed with cxcipients which are pharmaceutically acceptable and compatible with the active ingredient.
  • Foi oral administration, the uridine and/or choline or their physiologically tolerated derivatives such as salts, esters, N- oxides, and the like are mixed with additives customary for this purpose, such as vehicles, stabilizers, or inert diluents, and converted by customary methods into suitable forms for administration, such as tablets, coated tablets, hard or soft gelatin capsules, aqueous, alcoholic or oily solutions.
  • the uridine and/or choline or their physiologically tolerated derivatives such as salts, esters, N-oxides, and the like are converted into a solution, suspension, or emulsion, if desired with the substances customary and suitable for this purpose, for example, solubilizers or-other.
  • An active component can be foimulated into the composition as neutralized pharmaceutically acceptable salt forms
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide or antibody molecule), which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed from the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like
  • the salts of the uridine and/or choline are pharmaceutically acceptable salts.
  • Other salts are, in one embodiment, useful in the preparation of the compounds according to the invention or of their pharmaceutically acceptable salts.
  • Suitable pharmaceutically acceptable salts of the compounds of this invention include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulphuric acid, methanesulphonic acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic: acid, oxalic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
  • a pharmaceutically acceptable acid such as hydrochloric acid, sulphuric acid, methanesulphonic acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic: acid, oxalic acid, citric acid, tartaric acid, carbonic acid or phospho
  • HPLC analysis was performed using a Beckman System Gold apparatus
  • a standard HPLC method for measuring nucleosides yields separate peaks for uridine and cytidine; however, a coincidence of the cytidine and tyrosine peaks precludes accurate measurement of cytidine levels, as shown for human plasma samples ( Figure 1 ).
  • Tyrosine is present in many biological fluids, e.g., plasma or cerebrospinal fluid (CSF)
  • CSF cerebrospinal fluid
  • a modified HPLC method was used which distinguished cytidine and tyrosine peaks, permitting accurate measurement of cytidine levels ( Figure 2).
  • Oral administration of UMP increases plasma uridine levels in humans
  • Plasma uridine levels were assayed as described in Example 1. Plasma uridine levels increased in response to oral UMP in a dose-dependent fashion, then returned to baseline levels within 8 hr ( Figure 3).
  • Oral administration of uridine or UMP increases plasma uridine levels in gerbils
  • EXAMPLE 4 Oral administration of uridine or UMP increases brain uridine levels in gerbils
  • Brains were quickly removed from the skull after decapitation, frozen on dry ice, homogenized in 80% methanol, centrifuged, lyophilized and analyzed as described for Example 3
  • Uridine is Readily Converted to Cvtidine in the Brain [00177]
  • gerbils were orally administered 250 mg/kg body weight uridine, and 60 min later plasma and brain levels of cytidine and uridine were assessed.
  • the fold-increases relative to control animals was calculated and are depicted in Figure 9A (plasma) and Figure 9B (brain).
  • the fold-increase of cytidine was normalized to the fold increase of uridine, which was arbitrarily set as 100%.
  • Uridine Increases Levels of the Phospholipid Precursor CDP-Choline in the Brain and in a Neural Cell Line
  • PC12 cells were maintained in Minimal Essential Medium (MEM; Invitrogen,
  • Rats were acclimated to the animal facility for more than 7 days before fed a control laboratory diet (Teklad Global 16% protein rodent diet, TD.00217, Harlan Teldad, Madison, WI), or this diet fortified with UMP « 2Na + (2.5%, TD.03398, UMP « 2Na + ; Numico Research, the Netherlands) for 6 weeks.
  • a control laboratory diet Teklad Global 16% protein rodent diet, TD.00217, Harlan Teldad, Madison, WI
  • UMP « 2Na + (2.5%, TD.03398, UMP « 2Na + ; Numico Research, the Netherlands
  • HVA serotonin
  • 5-hydroxyindoleacetic acid 5-HIAA
  • DHBA 3,4-dihydroxybenzoic acid
  • Sigma Sigma (St Louis, MO) and were dissolved in HCIO 4 (0.1 M) to make 1 mM stock solutions, and aliquots were kept at -8O 0 C.
  • Ketamine hydrochloride 100 mg/ml was purchased from Fort Dodge Animal Health (Fort Dorge, IA).
  • Xylazine (20 mg/ml) originated from Phoenix Scientific, Inc. (St. Joseph, MO).
  • Ringer solution consisted of NaCl 147, KCl 2.7, CaCl 2 1.2 and MgCl 2 0 85 mM.
  • KCl was increased to 80 mM, with NaCl decreased to 69 7 mM to maintain osmolarity. All solutions were made from doubly distilled deionized water and filtered by Steriflip ® (Millipore, Bedford, MA)
  • Rats were anesthetized with a mixture of ketamine and xylazine (80 and 10 mg/Kg of body weight, respectively, intraperitoneally), and were placed in a Kopf stereotaxic frame All surgical instruments were sterilized by a hot bead dry sterilizer or 70% ethanol. A small hole was drilled into the skull by a 2-mm trephine bone drill. CMA/1 1 14/04 Cupr piobe (O.D.
  • the freely moving rat was perfused in a circular bowl on a rotating platform obviating the need for a liquid swivel (see Wang L el al, Neurochem Int 42: 465-70, 2003), and was habituated to the environment on the first day after surgery. Experiments were performed approximately 48 Iir after the surgery, and were carried out between 10:00 am to 4:00 pm. Ringer's solution was perfused continuously using Fluorinatedelhylenepropylene (FEP) Resin tubing and a gas-tight syringe (Exmire type I, CMA), at a constant rate of 1 5 ⁇ l/min by a microinfusion pump (CMA/100). Dialysates were collected at 15-min intervals.
  • FEP Fluorinatedelhylenepropylene
  • the mobile phase (MD- TM, ESA) consisted of 75 mM NaH 2 PO 4 , 1 7mM 1 -octanesulfonic acid, 100 ⁇ l/L Triethylamine, 25 ⁇ M EDTA, 10% acetonitrile, pH 3.0. The flow rate was 0.4 mL/min.
  • the column (ESA MD 150, 3 X 150 mm, 3 ⁇ m, 120 A) was kept in a 40 0 C column oven Samples were injected to HPLC by an ⁇ lltech 580 autosamplei (Alltech, Deerfield, IL) and maintained to 4°C with a cooling tray during analysis Data were captured by Alltech AllChiom " data system, and analyzed with AllCluom plus "' software A timeline program, which could change the detection gain during sample separation and detection, was used to make it possible to get low DA and high metabolites concentration data in dialysate through one injection.
  • ⁇ lltech 580 autosamplei Alltech, Deerfield, IL
  • Striatal tissues were placed in Eppendorf tubes containing 200 ⁇ l lysis buffer (60 mM Tris-HCl, 4% SDS, 20% glycerol, 1 mM dithiothreitol, 1 mM AEBSF, 8 ⁇ M aprotinin, 500 ⁇ M bestatin, 15 ⁇ M E64, 200 ⁇ M leupeptin, 10 ⁇ M pepstatin A).
  • the samples were sonicated, boiled (10 min), and centrifuged (14,000 g for 1 min at room temperature). The supernatant fluid was transferred to a clean tube, and total protein content was determined using the Bicinchoninic Acid assay (Sigma, St Louis, MO).
  • Equal amounts, of protein 40 ⁇ g protein/lane) were loaded /or sodium jdodecyl sulfate-polyacrylamide gel electrophoresis (4-15% SDS PAGE; Bio-Rad, Hercules, CA) Prior to gel electrophoresis, bromphenol blue solution (0.07%) was added to each sample Proteins were separated, transferred onto polyvinylidene difluoride (PVDF) membranes (Immobilon-P, Millipore), and blocked with 5% bovine serum albumin (Tris-buffered saline/0.15% Tween 20) for 1 h.
  • PVDF polyvinylidene difluoride
  • blots were incubated in TBST with various antibodies against the proteins of interest, including NF-70, NF-M (1 : 2000, 1 : 5000, respectively; Calbiochem, La .lolla, CA) at 4 °C overnight on an orbital shaker. Protein- antibody complexes were detected and visualized using the ECL system (Amersham, Piscataway, NJ) and Kodak X-AR film, respectively, as suggested by the manufacturer. Films were digitized using a Supervista S-12 scanner with a transparency adapter (UMAX Technologies, Freemont, CA). Analysis was performed using the public domain NIH Image program (NIH V.I 61).
  • NF-70 neurofilament-70
  • NF- M neurofilament-M
  • biomarkers of neurite outgrowth were assessed in the biains of the rats fiom the experiment described in Example 7
  • Uridine or UTP Increases Neurite Outgrowth and Branching
  • PC 12 cells were sparsely plated on collagen-coated 60 mm culture dishes in
  • Example 6 except that 5 mM NaH 2 PO,!, pH 2.65 was used as buffer A
  • Neurofilament proteins are highly enriched within neurites; ⁇ therefore, an ⁇ increase in neurile number should be associated with increased expression of neurofilament proteins NF-70 (70 kD) and NF-M (145 IcD) levels following 4-day treatment of PC 12 cells with NGF alone, or NGF plus uridine (50 ⁇ M) were thus measured ( Figure 16E). Both NF-70 and NF-M expression significantly (p ⁇ 0.01, p ⁇ 0.001 , respectively) increased following uridine treatment, compared Io cells treated only with NGF In the absence of NGF, uridine treatment had no effect on levels of either neurofilament protein. Thus, uridine augments neurite outgrowth in PC 12 cells
  • UTP or CTP levels in the presence of NGF were measured in PC 12 cells for 2 days with NGF, treated with no nucleotide, (control), uridine, cytidine or UTP, in the presence of NGF Uridine (50 ⁇ M) significantly (p ⁇ 0.05) increased both UTP and CTP levels
  • uridine or UTP dietary supplementation increased the levels of two major neurofilament proteins in rat brain, and was directly shown to induce neurite outgiowth in PC 12 cells.
  • NGF-differentiated PC 12 cells express pyrimidine-sensitive P2Y2, P2Y4 and P2Y6 receptors
  • P2Y2, anti-P2Y4 both from Calbiochem; or rabbit anti-P2Y6 (Novus Biologicals, Littleton. " CO).
  • PC 12 cells were treated as described above, except they were grown on 12mm glass cover slips (A Daigger & Co , Vernon Hills, IL) coated with collagen. Proteins were visualized using immunofluorescence. Briefly, the cells were fixed with 4% paraformaldehyde, permeabilized with 0.25% Triton X-I OO, blocked in 10% normal goat serum, and incubated overnight in the appropriate antibodies (mouse anti-NF-70, and either rabbit anti-P2Y2, rabbit anti-P2Y4 or rabbit anti-P2Y6). For P2Y2 and P2Y4 visualization, control cultures weie incubated with primary antibody plus a control antigen in order to ensure that the immuno- staining would be specific.
  • Control antigen was not available for the P2Y6 receptor Cells were then incubated in fluorochrome-conjugated secondary antibodies for 1 hour (goat anti-rabbit ALEXA 488 and goat anti-mouse ALEXA 568; Molecular Probes, Eugene, OR) and mounted on glass slides with mounting media with or without DAPI (Vector Laboratories, Burlingame, CA). Control antigens provided with the primary antibodies were used to ensure that immuno- staining was specific Digital images were obtained on a Zeiss (Oberkochen. Germany) Axioplan microscope with OpenLab software, using a Zeiss Plan-Neofluor 4Ox oil-immersion objective.
  • UTP is an agonist of the pyrimidine-activated class of P2Y receptors, namely
  • P2Y2, P2Y4 and P2Y6 receptors To determine whether these receptors participate in the mechanism by which extracellular UTP affects neuritc outgrowth, it was first determined whether the receptors are expressed in PC 12 cells, and whether exposure to NGF alters their expression, PC 12 cells were treated for 0 - 7 days with NGF and levels of the receptors measured. After 3 days of NGF treatment, expression of the P2Y2 receptor reached maximal levels, which were significantly (p ⁇ 0.001) higher than those seen at less than 3 days of NGF treatment (Figure 19A).
  • IP Phosphatidyl ⁇ ositol
  • P2Y2, P2Y4 and P2Y6 receptors activate the phospholipase
  • NGF-differentiated PC 12 cells were labeled with [3H]-inositoI (50 ⁇ M) or UTP (10,
  • IP signaling was assessed by measuring turnover of radio-labeled IP (Figure 22). Formation of IP was significantly incieased by addition of 100 ⁇ M UTP (p ⁇ 0.05) and by 50 ⁇ M uridine (p ⁇ 0.01 ). The P2Y receptor antagonist PPADS (100 ⁇ M) significantly (p ⁇ 0.05) blocked the stimulation of IP signaling by UTP.
  • the findings of Examples 10-12 provide a mechanism by which uridine and its metabolites stimulate neurite outgrowth: namely, by activation of P2Y receptors At least part of the action of the P2Y receptors is mediated by IP signaling
  • the findings from Examples 7-12 provide furthei evidence that uridine treatment can improve cognitive function by enhancing neurotransmission by multiple mechanisms: (1) enhancing neurotransmitter release; (2) acting, through CTP, as a precursor for membrane phosphatides; (3) activating, through UTP, the P2Y receptor-coupled intracellular signaling pathway Mechanisms (2) and (3) may act together to increase neurite formation.
  • UMP-supplemented diets enhance learning and memory in multiple species
  • UMP diets for six weeks. They were then shown a hidden platform in a six-foot diameter pool of water, placed somewhere in each of the four quadrants of the pool in turn, and were allowed 90 seconds in each trial to attempt to relocate the platform by swimming, and the swimming time "mean escape latency" recorded. The set of four trials was repeated on each of four consecutive days. The platform was in the same place each day. This test, known as the Morris water maze, is an indicator of spatial memory.
  • Working memory and reference memory assay Groups often gerbils fed control or 0.1 % UMP diet for four weeks and trained to successfully find all of the food pellets as described above were then given a modified test, in which only two arms of the maze (but always the same two) contained food pellet rewards. In this test, a working memory error is one in which a gerbil revisits an arm from which it has already taken the pellet that day. A reference memory error is one in which the gerbil enters an arm which never had food pellets (during the modified tests).
  • Brain slices are repeatedly stimulated as described in the previous Example, in this case for 8 cycles or alternating 20-minute periods of stimulation and rest
  • the amount of neurotransmitter release decreases with each successive stimulation period; however, this decrease is significantly less in the presence of either uridine or choline. This effect is enhanced by the presence of both uridine and choline.
  • uridine and choline the total amount of neurotransmitter release after repeated stimulation is increased by the presence of uridine or choline, and is further increased by the presence of uridine and choline.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Neurology (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Molecular Biology (AREA)
  • Psychiatry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Saccharide Compounds (AREA)

Abstract

The present invention is directed to methods of improving cognitive and neurological functions and increasing synthesis and release of neurotransmitters and membrane synthesis by neural cells and brain cells, comprising administering a composition comprising a uridine.

Description

COMPOSITIONS COlSfTAINING URIDINE, AND METHODS UTILIZING SAME
FIELD OF THE INVENTION
[0001 ] The present invention is directed to methods of improving cognitive and neurological functions and increasing synthesis and release of neurotransmitters and membrane synthesis by neural cells and brain cells, comprising administering a composition comprising a uridine or a source thereof.
BACKGROUND OF THE INVENTION
[0002] Uridine is a pyrimidine nucleoside and is essential in the synthesis of ribonucleic acids and tissue glycogens such as UDP glucose and UTP glucose. Prior medical uses of uridine alone include treatment of genetic disorders related to deficiencies of pyrimidine synthesis such as orotic aciduria. Choline, a dietary component of many foods, is part of several major phospholipids that are critical for normal membrane structure and function. Choline is included with lipid emulsions that deliver extra calories and essential fatty acids to patients receiving nutrition parenterally.
SUMMARY OF THE INVENTION
[0003] The present invention is directed to methods of improving cognitive and neurological functions and increasing synthesis and release of neuiotransmitters and membrane synthesis by neural cells and brain cells, comprising administering a composition comprising a uridine or a source thereof.
[0004] In one embodiment, the present invention provides a method of improving a cognitive function in a subject, comprising administering to the subject a uridine, a source thereof, or a composition comprising a uridine and a choline.
[0005] In another embodiment, the present invention provides a method of improving a neurological function in a subject, comprising administering to the subject a uridine, a source thereof, or a composition comprising a uridine and a choline.
[0006] In another embodiment, the present invention provides a method of treating or ameliorating a decline in a cognitive function in a subject, comprising administering a uridine, a source thereof, or a composition comprising a uridine and a choline to the subject. [0007] In another embodiment, the present invention provides a method of increasing or enhancing an ability of a brain cell or a neural cell of a subject to synthesize a neurotransmitter, comprising administering to the subject or the brain cell or neural cell a uridine, a source thereof, or a composition comprising a uridine and a choline
[0008] In another embodiment, the present invention provides a method of increasing a level of a neurotransmitter in a synapse, comprising contacting a neural cell adjacent to the synapse with a uridine, a source thereof, or a composition comprising a uridine and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a level of a neurotransmitter in a synapse.
[0009] In another embodiment, the present invention provides a method of increasing a level of a cytidine in a tissue, plasma, or cell of a subject, comprising administering a uridine, a source thereof, to the subject.
[0010] In another embodiment, the present invention provides a method of increasing a level of a cytidine in a tissue, plasma, or cell of a subject, comprising administering a composition comprising a uridine or a source thereof and a choline to the subject.
[0011] In another embodiment, the present invention provides a method of stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject, comprising contacting the subject with a uridine, a source thereof, or a composition comprising a uridine and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject.
[0012] In another embodiment, the present invention provides a method of stimulating or enhancing an outgrowth of a neurite of a neural cell, comprising contacting the neural cell with a uridine, a source thereof, or a composition comprising a uridine and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing an outgrowth of a neurite of a neural cell.
BRIEF DESCRIPTION OF THE DRAWINGS
[0013] Figure 1 illustrates the coincidence of cytidine and tyrosine peaks (6.59) when tested by a standard HPLC method. [0014] Figure 2 illustrates distinct cytidine (3.25) and tyrosine (2.92) peaks when tested by a modified HPLC method, which utilizes elution buffer with low methanol.
[0015] Figure 3 Oral UMP administration raises blood uridine levels in humans. Depicted is the ratio of uridine (set as 100% value) to cytidine in plasma after oral administration of 250 milligram per kg of body weight (mg/kg) of uridine.
[0016] Figure 4 Oral uridine administration raises blood uridine levels in gerbils. Depicted are plasma uridine levels 60 minutes following mock administration or administration of cytidine or uridine. **: p < 0.01 vs. mock-fed control; ##: p < 0.01 vs cytidine.
[0017] Figure 5. Oral UMP administration raises blood uridine levels in gerbils. Depicted are plasma uridine levels at various time points following administration or administration of water or UMP.
[0018] Figure 6. A UMP-supplemented diet raises blood uridine levels in gerbils. Depicted are plasma uridine levels in gerbils fed a diet containing the indicated percentages of UMP.
[0019] Figure 7. Oral uridine administration raises brain uridine levels. Depicted are brain uridine levels 60 minutes following mock administration or administration of cytidine or uridine. **: p < 0.01 vs. mock-fed control; ##: p < 0.01 vs cytidine.
[0020] Figure 8. Oral UMP administration raises brain uridine levels Depicted are brain uridine levels at various time points following administration or administration of water or UMP.
[0021] Figure 9 Uridine is readily converted to cytidine in the brain. Depicted is the ratio of uridine (100%) to cytidine in plasma (A) and in the brain (B) after oral administration of 250 milligram per kg of body weight (mg/kg) of uridine
[0022] Figure 10. Oral UMP administration raises brain CDP-choline levels. Depicted are brain CDP-choline levels at various time points following administration or administration of water or UMP.
[0023] Figure 1 1. Uridine increases intracellular levels of CDP-choline in a neural cell line. Cells were incubated for 6 h with the indicated concentrations of uridine. Depicted are the means +/- S.E.M. of six dishes, expressed as picomole (pmol) CDP-choline/mg protein. The experiment was repeated 3 times. *: p < 0 05 [0024] Figure 12. UMP dietary supplementation significantly increases potassium-evoked dopamine (DA) release in striatal dialysate. (A) Effect of dietary UMP supplementations on K+- evoked striatal DA release. Data were calculated from six to nine measurements at each point (means ± standard error of measurement [S.E.M.]) The 100% value represented the mean of the four measurements before potassium stimulation was set at 100%. (B) Data were pooled according to UMP treatment groups. "*" denotes p < 0 05 compared to corresponding contiols
[0025] Figure 13 Effect of potassium on DOPAC and HVA levels in striatal dialysate with UMP dietary supplementation. (A): DOPAC (B): HVA. *: p < 0.05 compared to corresponding controls
[0026] Figure 14. Increased acetylcholine basal concentration with UMP treatment. Depicted are means +/- SEM, "*" denotes p value of >0.05.
[0027] Figure 15. Effect of UMP dietary supplemention on neurofilament protein levels in contralateral striatum. (A): NF-70. (B): NF-M *: p < 0,05, **: p < 0 01 compared to corresponding controls.
[0028] Figure 16. Uridine treatment enhanced neurite outgrowth in PC 12 cells. A PC 12 cells treated for 4 days with NGF (50 ng/ml) in the presence or absence of uridine (50 μM) B. Number of neurites per cell after 2 or 4 days of treatment. C Number of neuiites per cell after 2 or 4 days of NGF plus different concentrations of uridine (50, 100 and 200 μM). D. Quantification of the number of branch points for each cell. E. Levels of the structural proteins NF-70 and NF-M, as determined using Western blotting. N = NGF, U = Uridine. Values represent means + SEM. **: p < 0 01, *** : p < 0.001 vs. NGF treatment.
[0029] Figure 17. Uridine treatment increased intracellular levels of UTP and CTP in PC 12 cells exposed to NGF for 2 days. Uridine treatment (50 μM) significantly increased intracellular UTP levels (A) and intracellular CTP levels (B) N - NGF, U = Uridine. C = Cytidine Values represent means + SEM * : p < 0.05 vs. NGF treatment
[0030] Figure 18. UTP treatment increased neurite outgrowth Treatment of PC 12 cells for 4 days with NGF and UTP significantly enhanced the number of neurites produced per cell, compared to treatment with NGF alone. Values represent means + SEM. **p < 0.01.
[0031 ] Figure 19. NGF-differentiated PC 12 cells express pyrimidine-sensitive P2Y receptors A Levels of P2Y2, P2Y4 and P2Y6 receptor expression after incubation of cells with NGF for varying lengths of time. B. Following 4 days of NGF treatment, cells were fixed and NF-70 (red) and P2Y receptor (green) proteins were visualized using immunofluorescence. Left panel: P2Y2. Middle panel: P2Y4, Right panel: P2Y6. Values represent means + SEM. *** p < 0.001
[0032] Figure 20. P2Y2 receptor co-localizes with the neuronal marker MAP-2. Left panel: P2Y2 receptor. Middle panel: MAP-2. Right Panel: Merge.
[0033] Figure 21. P2Y receptor antagonists inhibited the effect of uridine on neurite outgrowth. Cells were treated foi 4 days with NGF and with or without uridine (100 μM) and the P2Y receptoi antagonists PPADS, suramin, or RB-2. Values represent means + SEM. ***p < 0.001 vs. NGF treatment; Up < 0.05, ###p < 0 001 vs NGF plus uridine treatment.
[0034] Figure 22. Phosphatidylinositol (Pl) turnover is stimulated by UTP and uridine. Cells were metabolically labeled with [3H] inositol overnight, stimulated with UTP, uridine, or UTP plus PPADS in the presence of lithium at the indicated concentrations, and radio-labeled inositol phosphates derived from PI breakdown were measured by scintillation counting. Values represent means + SEM. *p < 0-05, **p < 0,01 vs. control; #p < 0.05 vs. 100 μM UTP treatment.
[0035] Figure 23 Oral UMP improves learning and spatial memory in rats. 18-month old rats in restricted environments consumed a control diet or a UMP diet for 6 weeks, and then were tested, using a Morris Water Maze, 4 trials/day for 4 days. Mean time to locate the platform is given in seconds.
[0036] Figure 24. Oral UMP improves learning and spatial memory in gerbils Learning and spatial memory of gerbils fed a control diet or diets containing the indicated amount of UMP were tested in a radial arm maze. Results are depicted as the amount of time remaining before the 3-minute deadline.
[0037] Figure 25. Oral UMP improves working memory and reference memory. The memory of gerbils fed a control or a 0.1% UMP diet for four weeks was tested using modification of the test depicted in Figure 24, which measured both working memory errors (A) and reference memory errors (B) Diamonds represent data points from control gerbils; triangles represent data points from gerbils fed 0.1% UMP diet [0038] Figure 26. Uridine and choline increase neurotransmitter release in striatal slices (top panel), hippocampal slices (middle panel), and cortical slices (top panel). Data are expressed as nanomoles per milligram protein per two hour, and depicted as means ± SEM. "*" = P < 0.001 relative to values obtained in the absence of choline. The first series in each panel was performed in the absence of choline; the second series was performed in the presence of choline. The bars in each series represent, from left to right, no additional compound added; cytidine added; and uridine added (each in addition to the choline, where appropriate).
DETAILED DESCRIPTION OF THE INVENTION
[0039] The present invention is directed to methods of improving cognitive and neurological functions and increasing synthesis and release of neurotransmitters and membrane synthesis by neural cells and brain cells, comprising administering a composition comprising a uridine or a source thereof.
[0040] In one embodiment, the present invention provides a method of improving a cognitive function in a subject, comprising administering to the subject a uridine or a source thereof, thereby improving a cognitive function in a subject
[0041 ] In one embodiment, the present invention provides a method of improving a cognitive function in a subject, comprising administering to the subject a composition comprising a uridine or a source thereof and a choline, thereby improving a cognitive function in a subject.
[0042] The phrase "uridine or a source thereof and a choline" refers to 2 embodiments of the present invention: a) a combination of uridine and choline; b) a combination of a uridine source and choline. The terms "uridine," "choline," and "uridine source" refer to any of their respective meanings that are mentioned herein. Each possibility represents a separate embodiment of the present invention.
[0043] In one embodiment, the cognitive function is memory. The memory is, in other embodiments, spatial memory, working memory, reference memory, short-term memory, long- term memory, or medium-term memory. In aΕ6tHeFembo"dimeήf,' the memόry i∑fany dtHeFtype" of memory known in the art. Each type of memory represents a separate embodiment of the present invention.
[0044] As provided herein, the data in Figures 21-23 show directly that uridine improves several types of memory. The consistency of the effect across different species in different types of assessments of memory verifies the findings of the present invention The data in Example 15 further show that the effects of uridine are enhanced by inclusion of a choline. Thus, administration of compositions comprising uridine and choline are effective at improving memory- more effective, in one embodiment, than administration of either uridine or choline alone.
[0045] In another embodiment, the cognitive function is learning The learning is, in other embodiments, cognitive learning, affective learning, or psychomotor learning. In another embodiment, the learning is any other type of learning known in the art. Each type of learning represents a separate embodiment of the present invention.
[0046] In another embodiment, the cognitive function is intelligence In other embodiments, the intelligence is linguistic intelligence, musical intelligence, spatial intelligence, bodily intelligence, interpersonal intelligence, intrapersonal intelligence, interpersonal intelligence, or logico-mathematical intelligence. In another embodiment, the intelligence is any other type of intelligence known in the art Each type of intelligence represents a separate embodiment of the present invention
[0047] In another embodiment, the cognitive function is mental fitness. In another embodiment, the cognitive function is any other type of cognitive function known in the art. Each type of cognitive function represents a separate embodiment of the present invention.
[0048] In one embodiment, "improving'" a cognitive function, or "improvement" of a cognitive function refer to increasing the capacity of the subject to perform the cognitive function. In another embodiment, the terms refer to an increased or improved baseline level of the cognitive fuηction in the subject. In another embodiment, the terms refer to an increased or improved level of the cognitive function in response to a challenge or test.
[0049] In another embodiment, improving a cognitive function refers to effecting a 10% improvement thereof In another embodiment, a 20% improvement is attained In other embodiments, a 30% improvement, a 40% improvement, a 50% improvement, a 60% improvement, a 70% improvement, an 80% improvement, or a 90% improvement is attained. In another embodiment, improving a cognitive function refers to effecting a 100% improvement thereof Each possibility represents a separate embodiment of the present invention. [0050] In another embodiment, improvement of a cognitive function is assessed relative to the cognitive function before beginning treatment. In anothei embodiment, improvement of a cognitive function is assessed relative to an untreated subject. In another embodiment, improvement of a cognitive function is assessed according to a standardized criterion such as, for example, a test or the like. Each type of improvement of cognitive activity represents a separate embodiment of the present invention
[0051] In another embodiment, improvement of a cognitive function is assessed by the number of connections between neurons in the subject's brain. In another embodiment, the improvement is assessed by the number of capillaries in the subject's brain, or in a specific region of the subject's brain. In another embodiment, the improvement is assessed by neural activity. In other embodiments, the improvement is assessed by neural function, linguistic function, or ability to communicate In another embodiment, the improvement is assessed by measurement of levels of acetylcholine or other neurotransmitters or brain chemicals coπelated with cognitive function. In other embodiments, the improvement is assessed by Positron Emission Tomography (PET) scanning of the subject's brain, magnetic resonance imaging (MR!) scanning of the subject's brain. In another embodiment the improvement is assessed by Cognitive Abilities Screening Instrument (CASI) (Peila R et al, Stroke. 32: 2882-9, 2001). In another embodiment, the improvement is assessed by a test such as, for example, the tests disclosed herein (Example 13). Additional methods for assessing improvement of cognitive function are well known in the art, and are described, for example in Antonova E et al (Schizophr Res. 2004 Oct l ;70(2-3):117-45) and in Cognitive Function Analysis (Greenwood Pub Group, 1998). Each method represents a separate embodiment of the present invention
[0052] In one embodiment of methods of the present invention, a composition of the present invention increases a level of cytidine, in the subject, thereby mediating one of the effects described herein (e.g improving cognitive or neurological function, stimulating neural function, membrane synthesis, neurotransmitter release, etc). In another embodiment, the effect is mediated by increasing a level of cytidine triphosphate (CTP) in the subject. In another embodiment, the effect is mediated -by increasing a level of CDP-choline in the subject In another embodiment, the effect is mediated by increasing a level of a derivative of cytidine, CTP, CDP-choline in the subject. In another embodiment, the effect is mediated by increasing a level of a metabolite of cytidine, CTP, CDP-choline in the subject. In another embodiment, the effect is mediated without increasing a level of cytidine, CTP, CDP-choline, or a derivative or metabolite thereof. Each possibility represents a separate embodiment of the present invention. Each possibility represents a separate embodiment of the present invention.
[0053] As described herein, Figures 9-1 1 show that orally administered uridine acts rapidly and effectively to raise levels of cytidine in the brain. In combination with Figures 3-8, which show that uridine is effectively and rapidly absorbed into the bloodstream, in several species, including humans, these findings demonstrate that administration of uridine raises levels of cytidine, CTP, and CDP-choline. The data in Example 15 further show that the effects of uridine are enhanced by inclusion of a choline.
[0054] In one embodiment, the cytidine level is a systemic level. In another embodiment, the cytidine level is a brain level In another embodiment, the cytidine level is a nervous system level. Each possibility represents a separate embodiment of the present invention.
[0055] In another embodiment, the potential benefit of uridine administration is greater than the benefit of cytidine administration This is due to the fact that cytidine, as opposed to uridine, either cannot cross or is much less efficient than uridine in crossing the blood-brain barrier (Cornford et al., Independent blood-brain barrier transport systems for nucleic acid precursors. Biochim. Biophys. Acta 349:21 1-219, 1975).
[0056] In another embodiment, the increase in cytidine, CTP, or CDP-choline or a derivative or metabolite thereof enables the cell to increase levels of a phospholipid, thereby mediating one of the effects described herein. In one embodiment, the phospholipid is phosphatidylcholine (PC) In another embodiment, the phospholipid is phosphatidylethanolamine (PE). In another embodiment, the phospholipid is phosphatidylserine (PS). In another embodiment, the phospholipid is or a derivative or metabolite of PC, PE, oi PS Each possibility represents a separate embodiment of the present invention.
[0057] In another embodiment, the present invention provides a method of improving a neurological function in a subject, comprising administering to the subject a uridine or a source thereof, thereby improving a neurological function in a subject.
[0058] In another embodiment, the present invention provides a method of improving a neurological function in a subject, comprising administering to the subject a composition comprising a uridine or a source thereof and a choline, thereby improving a neurological function in a subject. [0059] In another embodiment, the neurological function that is improved by a method of the present invention is a synaptic transmission. In one embodiment, the synaptic transmission is adjacent to a motor neuron. In another embodiment, the synaptic transmission is adjacent to an intemeuron. In one embodiment, the synaptic transmission is adjacent to a sensory neuron Each type of synaptic transmission represents a separate embodiment of the present invention.
[0060] In anothei embodiment, the synaptic transmission is improved or enhanced by means of stimulating oi enhancing an outgrowth of a neurite of a neural cell. In another embodiment, stimulating or enhancing an outgrowth of a neurite of a neural cell is partially responsible for improving or enhancing the synaptic transmission. In another embodiment, a composition of the present invention improves or enhances synaptic transmission without stimulating an outgrowth of a neurite. Each possibility represents a separate embodiment of the present invention.
[0061] "Neurite" refers, in one embodiment, to a process growing out of a neuron In one embodiment, the process is a dendrite. In another embodiment, the process is an axon Each type of neurite represents a separate embodiment of the present invention.
[0062] In another embodiment, the synaptic transmission is improved or enhanced by increasing the number of neurites of the neural cell. In another embodiment, improvement or enhancement of the synaptic transmission occurs without increasing the number of neurites of the neural cell Each possibility represents a separate embodiment of the present invention
[0063] In another embodiment, the synaptic transmission is improved oi enhanced by stimulating or enhancing blanching of a neurite of a neural cell In another embodiment, improvement or enhancement of the synaptic transmission occurs without stimulating or enhancing branching of a neurite of a neural cell Each possibility represents a separate embodiment of the present invention.
[0064] The data of Example 9 shows that when levels of membrane precursors are increased, neurons produce moie neurites, with more branches. By increasing its surface aiea and size, a cell is able, in one embodiment, to form more connections with neighboring cells. Moreover, an increase in the amount or composition of plasma membiane alters, in one embodiment, neurotransmitter synthesis and release, which also, in one embodiment, affects memory formation. Thus, compounds that promote neurite outgrowth, such as uridine, are useful for treatment of neurodegenerative disorders like Alzheimer's disease, which involves loss of neuronal connections and memory impairment. [0065] In anothei embodiment, improving the synaptic transmission in the subject is achieved by increasing an amount of a membrane of a neural cell as a result of administration of the uridine and/or choline. In another embodiment, the improvement is achieved by stimulating a synthesis of a membrane of a neural cell. In another embodiment, the improvement is achieved by enhancing a synthesis of a membrane of a neural cell. In another embodiment, stimulating or enhancing an amount of or a synthesis of a membrane of a neural cell is partially responsible for mediating improving the synaptic transmission in the subject. In another embodiment, the uridine and/or choline improves the synaptic transmission without stimulating or enhancing an amount of or a synthesis of a membrane of a neural cell. Each possibility represents a separate embodiment of the present invention
[0066] In another embodiment, the neurological function that is improved or enhanced is a function of a neurotransmitter. In one embodiment, the improvement occurs by means of increasing a level of the neurotransmitter in a synapse. In another embodiment, the improvement occurs by means of increasing the release of the neurotransmitter into a synapse. In another embodiment, the improvement occurs without changing the level or release of the neurotransmitter in a synapse. Each possibility represents a separate embodiment of the present invention.
[0067] As provided herein, the data in Figures 12-13 show that uridine significantly improves neurotransmitter function, highlighting the ability of uridine to improve neurological function. The data in Figures 14-17 show a beneficial effect of uridine on the morphology of neurites, further demonstrating the ability of uridine to improve neurological function The data in Example 15 further show that the effects of uridine are enhanced by inclusion of a choline Thus, administration of compositions comprising uridine and choline are effective at improving neurological function - more effective, in one embodiment, than administration of either uridine or choline alone.
[0068] In another embodiment, the present invention provides a method of treating or ameliorating a decline in a cognitive function in a subject, comprising administering a uridine or a source thereof to the subject, thereby treating or ameliorating a decline in a cognitive function in a subject.
[0069] In another embodiment, the present invention provides a method of treating or ameliorating a decline in a cognitive function in a subject, comprising administering a composition comprising a uridine or a source thereof and a choline to the subject, thereby inhibiting or preventing a decline in a cognitive function in a subject
[0070] "Treating or ameliorating a decline in a cognitive function" refers, in one embodiment, to mitigating the decline In another embodiment, the phrase refers to preventing the decline. In another embodiment, the phrase refers to ieversing the decline. In another embodiment, the phrase refers to slowing the decline. In another embodiment, the phrase refers to halting the decline Each possibility represents a separate embodiment of the present invention
[0071] In another embodiment, the decline in a cognitive function results from a neurological disorder. In one embodiment, the neurological disorder is a memory disorder The memory disorder comprises, in one embodiment, a memory decline. In another embodiment, the memory decline is associated with brain aging. In other embodiments, the memory disorder is selected from Pick's disease, Lewy Body disease, or a dementia. In other embodiments, the dementia is associated with Huntington's disease or AIDS dementia. Each possibility represents a separate embodiment of the present invention.
[0072] In another embodiment, the decline in a cognitive function results from a neurodegenerative disease. In one embodiment, the neurodegenerative disease is Alzheimer's disease. In other embodiments, the neurodegenerative disease is amyotrophic lateral sclerosis, multiple system atrophy, Parkinson's disease, progressive supranuclear palsy, frontotemporal dementia, Huntington's disease, or a prion disease. In another embodiment, the neurodegenerative disease is any other neurodegenerative disease known in the art. Eacli possibility represents a separate embodiment of the present invention
[0073] In another embodiment, the decline in a cognitive function results from a cardiovascular disease In one embodiment, the cardiovascular disease is a stroke In another embodiment, the cardiovascular disease is a multi-infarct dementia. In another embodiment, the cardiovascular disease is any other cardiovascular disease known in the art. Each possibility represents a separate embodiment of the present invention.
[0074] In one embodiment, the neurological disorder is associated with a dopaminergic pathway In another embodiment, the neurological disorder is not associated with a dopaminergic pathway Each possibility represents a separate embodiment of the present invention. [0075] In another embodiment, the neurological disorder is a cognitive dysfunction. In one embodiment, the cognitive dysfunction is dyslexia. In other embodiments, the cognitive dysfunction comprises a lack of attention, a lack of alertness, a lack of concentration, or a lack of focus. In other embodiments, the cognitive dysfunction comprises minimal cognitive impairment or age-ielated memory impairment. Each possibility represents a separate embodiment of the present invention.
[0076] In another embodiment, the neurological disorder is an emotional disorder. In other embodiments, the emotional disorder comprises mania, depression, stress, panic, anxiety, dysthymia, or psychosis. In another embodiment, the emotional disorder comprises a seasonal effective disorder. In another embodiment, the emotional disorder comprises a bipolar disorder.
[0077] In another embodiment, the neurological disorder is a psychiatric disease. In another embodiment, the neurological disorder is a depression. In one embodiment, the depression is an endogenous depression. In another embodiment, the depression is a major depressive disorder. In another embodiment, the depression is depression with anxiety. In another embodiment, the depression is bipolar depression. Each type of depression iepiesents a separate embodiment of the present invention.
[0078] In another embodiment, the neurological disorder is selected from the group consisting of ataxia and Friedreich's ataxia
[0079] In another embodiment, the neurological disorder is a movement disorder. The movement disorder comprises, in other embodiments, a tardive dyskinesia, a dystonia, or a Tourette's syndrome. In another embodiment, the movement disorder is any other movement disorder known in the art
[0080] In another embodiment, the neurological disorder is a cerebrovascular disease. The cerebro-vascular disease results, in one embodiment, from hypoxia In another embodiment, the disease results from any other cause capable of causing a cerebro-vascular disease. In another embodiment, the disease is cerebral thrombosis. In another embodiment, the cerebro-vascular disease is ischemia.
[0081 ] In another embodiment, the neurological disorder is a behavioral syndrome. In another embodiment, the neurological disorder is a neurological syndrome. In other embodiments, the behavioral syndrome or neurological syndrome follows brain trauma, spinal cord injury, or anoxia.
[0082] In another embodiment, the neurological disorder is a peripheral nervous system disorder. In other embodiments, the peripheral nervous system disorder is a neuromuscular disorder, myasthenia gravis, or post-polio syndrome. In another embodiment, the peripheral nervous system disorder is any other peripheral nervous system disorder known in the art In another embodiment, the neuromuscular disorder is a muscular dystrophy
[0083] Each type of neurological disorder mentioned herein represents a separate embodiment of the present invention.
[0084] In another embodiment, the present invention provides a method of increasing or enhancing an ability of a brain cell or a neuial cell of a subject to synthesize a neurotransmitter, comprising administering to the subject or the brain cell or neural cell a uridine or a source thereof, thereby increasing or enhancing an ability of a brain cell of a subject to synthesize a neurotransmitter.
[0085] In another embodiment, the present invention provides a method of increasing or enhancing an ability of a brain cell or a neural cell of a subject to synthesize a neurotransmitter, comprising administering to the subject or the brain cell or neural cell a composition comprising a uridine or a source thereof and a choline, thereby increasing or enhancing an ability of a brain cell of a subject to synthesize a neurotransmitter.
[0086] In another embodiment, the present invention provides a method of increasing or enhancing an ability of a brain cell or a neural cell of a subject to repeatedly release an effective quantity of a neurotransmitter into a synapse, comprising administering to the subject or the brain cell or neural cell with a uridine or a source thereof, thereby increasing or enhancing an ability of a brain cell or a neural cell of a subject to repeatedly release an effective quantity of a neurotransmitter into a synapse.
[0087] In another" embodiment", th"e "present" invention" provides a "method of increasing" of " enhancing an ability of a brain eel) or a neural cell of a subject to repeatedly release an effective quantity of a neurotransmitter into a synapse, comprising administering to the subject or the brain cell or neural cell with a composition comprising a uridine or a source thereof and a choline, thereby increasing or enhancing an ability of a brain cell or a neural cell of a subject to repeatedly release an effective quantity of a neurotransmitter into a synapse, As described herein, findings of the present invention show that uridine enhances the ability of neurons to synthesize neurotransmitters and repeatedly release them (Example 7) The data in Example 15 further show that this effect of uridine is enhanced by inclusion of choline.
[0088] In one embodiment, the release which is enhanced by a method of the present invention occurs following a stimulation of the neuron. In one embodiment, the release which is enhanced occurs following a depolarization of the neuron. In one embodiment, the release which is enhanced is a basal neurotransmitter release. In one embodiment, the stimulation of the neuron comprises exposure of the neuron to a potassium ion. In another embodiment, the stimulation of the neuron comprises any other means of neural stimulation known in the art. Methods for assessing neural stimulation and release of neurotransmitters are well known in the art, and are described, for example, in Bewick GS, J Neurocytol 32: 473-87, 2003. Each possibility represents a separate embodiment of the present invention
[0089] In another embodiment, the picsent invention provides a method of increasing a level of a neurotransmitter in a synapse, comprising contacting a neural cell adjacent to the synapse with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a level of a neurotransmitter in a synapse.
[0090] In another embodiment, the present invention provides a method of increasing a level of a neurotransmitter in a synapse, comprising contacting a neural cell adjacent to the synapse with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a level of a neurotransmitter in a synapse.
[0091] In another embodiment, the present invention provides a method of increasing a sensitivity of a neuron to a stimulus, comprising contacting the neuron with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a sensitivity of a neuron to a stimulus,
[0092] In another embodiment, the present invention provides a method of increasing a sensitivity of a neuron to a stimulus, comprising contacting the neuron with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a sensitivity of a neuron to a stimulus. [0093] In one embodiment, the neurotransmitter whose levels or activity, or release is affected by methods of the present invention is acetylcholine. In another embodiment, the neurotransmitter is dopamine In another embodiment, the neurotransmitter is serotonin. In another embodiment, the neurotransmitter is 5-hydroxytryptamine (5-HT). In another embodiment, the neurotransmitter is GABA. In another embodiment, the neurotransmitter is any other neurotransmitter known in the art. Each type of neurotransmitter represents a separate embodiment of the present invention
[0094] In another embodiment, the present invention provides a method of stimulating a production of a phosphatidylcholine (PC) by a brain cell or neural cell of a subject, compiising administering to the subject or brain cell or neural cell a uridine or a source thereof, thereby stimulating a production of a PC by a brain cell or neural cell.
[0095] In another embodiment, the present invention provides a method of stimulating a production of a phosphatidylcholine (PC) by a brain cell or neural cell of a subject, comprising administering to the subject or brain cell or neural cell a composition comprising a uridine or a source thereof and a choline, thereby stimulating a production of a PC by a brain cell or neural cell. As described herein, findings of the present invention show that uridine enhances synthesis of the PC precursor CDP-choline (Example 6). The data in Example 15 further show that this effect of uridine is enhanced by inclusion of choline.
[0096] In another embodiment, the present invention provides a method of stimulating or enhancing an amount of or a synthesis of a component of a cell membrane, comprising contacting the cell with a uridine or a source theieof, thereby stimulating or enhancing an amount of or a synthesis of a cell membrane.
[0097] In another embodiment, the present invention provides a method of stimulating or enhancing an amount of or a synthesis of a component of a cell membrane, comprising contacting the cell with a composition comprising a uridine or a source thereof and a choline, thereby stimulating or enhancing an amount of or a synthesis of a cell membrane.
[0098] In another embodiment, the component whose synthesis is enhanced by a method of the present invention is a PC. In another embodiment, the component is a glyceiophospholipid. In another embodiment, the component is a phosphatide acid In another embodiment, the component is a PE In another embodiment, the component is a lecithin. In anothei embodiment, the component is a Pl. In another embodiment, the component is a PS In other embodiments, the component is a 2-lysolecithin, a plasmalogen, a choline plasmalogen, a phosphatidylglycerol, a choline diphosphatidylglycerol, a choline sphingolipid, or a choline sphingomyelin. In another embodiment, the component is any other phospholipid known in the art Each type of phospholipid represents a separate embodiment of the present invention.
[0099] In another embodiment, the present invention provides a method of stimulating or enhancing an amount of or a synthesis of a phospholipid precursor, comprising contacting the cell with a uridine or a source thereof, thereby stimulating or enhancing an amount of or a synthesis of a phospholipid precursor.
[00100] In another embodiment, the present invention provides a method of stimulating or enhancing an amount of or a synthesis of a phospholipid precursor, comprising contacting the cell with a composition comprising a uridine or a source thereof and a choline, thereby stimulating or enhancing an amount of or a synthesis of a phospholipid precursor. In another embodiment, the phospholipid precursor is CDP-choline (Example 6). In another embodiment, the phospholipid precursor is CTP In another embodiment, the phospholipid precursor is inositol. In another embodiment, the phospholipid precursor is choline In another embodiment, the phospholipid precursor is glycerol. In another embodiment, the phospholipid precursor is acetate In anothei embodiment, the phospholipid precursor is any other phospholipid precursor known in the art. Each phospholipid precursor represents a separate embodiment of the present invention.
[00101] In another embodiment, the present invention provides a method of stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject, comprising contacting the subject with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject
[00102] In another embodiment, the present invention provides a method of stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject, coiήprising"contacting the subject with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing a production of a membrane of a brain cell or a neural cell of a subject [00103] In one embodiment, the membrane is a neurite membrane. In another embodiment, the membrane is a dendritic membrane. In another embodiment, the membrane is an axonal membrane. In another embodiment, the membrane is any other type of membiane known in the art. Each type of membrane represents a separate embodiment of the present invention.
[00104] In another embodiment, stimulating an amount of or a synthesis of the cell membrane is accomplished by stimulating or enhancing a synthesis of a phospholipid (Example 6). In another embodiment, stimulating or enhancing an amount of or a synthesis of a membrane of a neural cell is accomplished by stimulating or enhancing a synthesis of a phospholipid precursor (Example 6) In another embodiment, stimulating oi enhancing a synthesis of a phospholipid or a precursor thereof is partially responsible for stimulating an amount of or a synthesis of a membrane of a neural cell In another embodiment, a composition of the present invention stimulates the amount of or a synthesis of a membrane without stimulating or enhancing a synthesis of a phospholipid or a precursor thereof Each possibility represents a separate embodiment of the present invention
[00105] Methods for assessing production of a brain cell membrane or neuial cell membrane are well known in art. In anothei embodiment, membrane production is assessed by measuring the level of neurite outgrowth or branching (Example 9). In anothei embodiment, membrane production is assessed by measuring the level of a membrane maikei protein (Example 8). In another embodiment, membrane production is assessed by measuring synthesis of a membrane precursor. In another embodiment, membrane production is assessed by measuring amounts of membrane prior to and following uridine treatment. In another embodiment, membrane production is assessed by measuring biological indicators of membrane turnover. Indicators or cellular membrane tumovei are well known in the art, and are described, for example, in Das KP et al, Neurotoxicol Teratol 26(3): 397-406, 2004 Each method of assessing membrane production represents a separate embodiment of the present invention.
[00106] In another embodiment, the present invention provides a method of stimulating or enhancing an outgrowth of a neurite of a neural cell, comprising contacting the neural cell with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing an outgrowth of a neurite of a neural cell [00107] In anolhei embodiment, the present invention provides a method of stimulating or enhancing an outgrowth of a neurite of a neural cell, comprising contacting the neural cell with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby stimulating or enhancing an outgrowth of a neurite of a neural cell. As described herein, findings of the present invention show that uridine enhances outgrowth and branching of neurites (Example 9). The data in Example 15 further show that this effect of uridine is enhanced by inclusion of choline.
[00108] In another embodiment, the present invention provides a method of increasing a number of neurites of a neuial cell, comprising contacting the neural cell with a uridine or a source thereof, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a number of neurites of a neural cell.
[00109] In another embodiment, the present invention provides a method of increasing a number of neurites of a neural cell, comprising contacting the neural cell with a composition comprising a uridine or a source thereof and a choline, whereby the composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a number of neurites of a neural cell
[001 10] In another embodiment, the present invention provides a method of stimulating or enhancing a branching of a neurite of a neural cell, comprising contacting the neural cell with a uridine or a source thereof, thereby stimulating or enhancing a branching of a neurite of a neural cell
[001 1 1 ] In another embodiment, the present invention provides a method of stimulating or enhancing a branching of a neurite of a neural cell, comprising contacting the neural cell with a composition comprising a uridine or a source thereof and/or a choline, thereby stimulating or enhancing a branching of a neurite of a neural cell
[001 12] In one embodiment, the cell that is the target of methods of the present invention or is contacted in the methods is a neural cell. In another embodiment, the cell is a brain cell In another embodiment, the cell is any cell in which synthesis of a membrane or a component thereof is enhanced by contact with a composition comprising a uridine and/or a choline. In another embodiment, the cell is any cell in which a neurological function is enhanced by contact with a composition comprising a uridine and/or a choline. Each possibility represents a separate embodiment of the present invention.
[001 13] - In another embodiment, the neural cell, neuiite, or brain cell of methods of the present invention is newly differentiated. In another embodiment, the cell is not newly differentiated. In one embodiment, "newly differentiated"' refers to a neuron that has differentiated in the 24 hours prior to commencing administration of the uridine and/or choline In another embodiment, the neuron has differentiated in the 48 hours prior to administration. In another embodiment, the neuron has differentiated in the 72 hours prior to administration In another embodiment, the neuron has differentiated in the 1 week prior to administration. In another embodiment, "newly differentiated"' refers to a neuion that completes its differentiation following commencement of administration of the composition of the present invention. Each possibility represents a separate embodiment of the present invention.
[001 14] Methods of assessing neuronal differentiation are well known in the art, and are described, for example, in Contestabile A et al (Neurochem Int 45: 903-14, 2004). Each such method represents a separate embodiment of the present invention.
[001 15] In another embodiment, the present invention provides a method of increasing a level of a cytidine in a tissue, plasma, or cell of a subject, comprising administering a uridine or a source thereof to the subject, thereby increasing a level of a cytidine in a tissue, plasma, or cell
[001 16] In another embodiment, the present invention provides a method of increasing a level of a cytidine in a tissue, plasma, or cell of a subject, comprising administering a composition comprising a uridine or a source thereof and a choline to the subject, thereby increasing a level of a cytidine in a tissue, plasma, or cell. In another embodiment, the piesent invention provides a method of increasing a level of a CTP in a tissue, plasma, or cell of a subject, comprising administering a composition of the present invention to the subject In another embodiment, the present invention provides a method of increasing a level of a CDP- cholme in a tissue, plasma; or ceil of a subject, compri sing-administering a composition of the present invention. In another embodiment the present invention provides a method of increasing a level of a derivative of a cytidine, a CTP, or a CDP-choline in a tissue, plasma, or cell of a subject, comprising administering a composition of the present invention In another embodiment, the present invention provides a method of increasing a level of a metabolite of a cytidine, a CTP, or a CDP-choline in a tissue, plasma, or cell of a subject, comprising administering a composition of the present invention. Each possibility represents a separate embodiment of the present invention
[00117] In one embodiment, the tissue is a brain tissue. In one embodiment, the tissue is a neural tissue. In another embodiment, the tissue is a spinal tissue. In another embodiment, the tissue is any other tissue known in the art.
[001 18] In one embodiment, the cell is a brain cell. In one embodiment, the cell is a neural cell. In another embodiment, the cell is a spinal cell. In another embodiment, the cell is any other cell known in the art. Each possibility represents a separate embodiment of the present invention.
[001 19] In one embodiment, the uridine that is administered in the present invention is a uridine-5'-monophosphate (UMP). In another embodiment, the uridine is a uridine-5"- diphosphate (UDP) In another embodiment, the uridine is a uridine-5 "-triphosphate (UTP). In another embodiment, the uridine is UDP glucose. Each possibility represents a separate embodiment of the present invention
[00120] In another embodiment, a uridine precursor is administered in methods of the present invention In one embodiment, the uridine precursor that is administered is a cytidine- 5'-monophosphate. In another embodiment, the uridine precursor that is administered is a cytidine-5' -diphosphate (CDP). In another embodiment, the uridine precursor that is administered is a CDP-glucose. In another embodiment, the uridine precursor that is administered is any pharmacologically acceptable uridine precursor, deiivative or metabolite known in the art.
[00121] In another embodiment, a uridine derivative is administered in methods of the present invention. The term "derivative" in one embodiment refers to a compound chemically related to uridine in such a way that uridine is converted to the derivative in a subject's body. In another embodiment, "derivative" refers to a compound chemically related to uridine in such a way that the derivative is converted to uridine in a subject's body In one embodiment, the conversion occurs via one or more stable intermediates. In another embodiment, the conversion occurs directly. Each possibility represents a separate embodiment of the present invention.
[00122] In another embodiment, a uridine metabolite is administered in methods of the present invention [00123] In other embodiments, uridine-based compounds other than uridine itself serve as uridine sources or uridine precursors. These are, in some embodiments, uridine-rich food or dietary products like algae; salts of uridine like uridine phosphates, acylated uridine or the like. In another embodiment, therapeutically or pharmacologically effective doses of acyl derivatives of uridine or mixtures theieof, e.g. those disclosed in U.S. Pat. No. 5,470,838, are administered.
[00124] In another embodiment, the uridine sourse is cytidine-diphosphocholine (CDP- choline; citicholine). While citicholine contains choline as well as uridine in a 1 :1 molar ratio, it is not, in one embodiment, sufficient to supply all the choline required by the subject. Thus, in this embodiment, citicholine serves a the source of all the uridine and some of the choline required by the subject.
[00125] In another embodiment, a salt of the uridine piecursor, derivative or source is utilized in a method of the present invention In one embodiment, the salt is UMP disodium (Examples 2-3) In another embodiment, the salt is any other pharmacologically acceptable salt of a uridine precursor or derivative. In another embodiment, the composition that is administered comprises the salt of the uiidine or precursor or derivative thereof as the sole active ingredient. Each uridine salt rcpiesents a separate embodiment of the present invention.
[00126] In another embodiment, a mixture of two oi more of the above uridine-related compounds is administered. Each type of uridine precursor, derivative, metabolite, or source represents a separate embodiment of the present invention.
[00127] The term "uridine'" as used herein refers, in one embodiment, to any uridine phosphate, uridine precursor, uridine metabolite, uridine-based compound, oi salt thereof mentioned above. In another embodiment, "uridine" refers to any uridine or related compound that is known in the art. Each possibility represents a separate embodiment of the piesent invention
[00128] In one embodiment, the uridine, derivative, source, or precursor thereof is administered in methods of the present invention in a dosage of between about 20 milligrams (mg) and 50 giams (g) per day. In another embodiment, the uridine or related compound is administered in a dosage of about 50 mg-30 g per day. In other embodiments, the dosage is about 75 mg-20 g; 100 mg-20 g; 100 mg-10 g; 200 mg-8 g; 400 mg-6 g; 600 mg-4 g; 800 mg-3 g; 1 -2.5 g; 1 5-2 g; 5 mg-5 g; or 5 mg-50 g per day. Each dosage or dosage range represents a separate embodiment of the present invention. [00129] In one embodiment, the choline administered in methods of the present invention is a choline salt. In one embodiment, the salt is choline chloride. In another embodiment, the salt is choline bitartrate. In another embodiment, the salt is choline stearate. In other embodiments, the salt is choline alfoscerate, choline dehydrocholate., choline dihydrogen citrate, or choline salicylate In another embodiment, the salt is any other choline salt known in the art. Each possibility represents a separate embodiment of the present invention
[00130] In another embodiment, the choline is a choline-based compound, e.g. a choline ester.
[00131 ] In another embodiment, the choline is a compound that dissociates to choline. In one embodiment, the compound is sphingomyelin In another embodiment, the compound is an acylglycerophosphocholine. In another embodiment, the compound is lecithin. In another embodiment, the compound is lysolecithin. In another embodiment, the compound is glycerophosphatidylcholine. In another embodiment a mixture of two or more of the above choline-related compounds is administered
[00132] The term "choline" as used herein refers, in one embodiment, to any choline phosphate, choline precursor, choline metabolite, choline-based compound, or salt thereof mentioned above In another embodiment, "choline" refers to any choline or related compound that is known in the art Each possibility represents a separate embodiment of the present invention
[00133] In another embodiment, the choline or choline-related compound is administered in such a manner and dosage that a choline level of at least 20-30 nanomoles is attained in the subject's blood or brain In another embodiment, a choline level of 10-50 nanomoles is attained. In another embodiment, a choline level of 5-75 nanomoles is attained. In another embodiment, a choline level of 25-40 nanomoles is attained. In another embodiment, a choline level of 30-35 nanomoles is attained. Each possibility represents a separate embodiment of the present invention.
[00134] In another embodiment, the choline, derivative, source, or precursor thereof is administered in methods of the present invention in a dosage of 20 mg-50 g per day In other embodiments, the choline or related compound is administered in a dosage of about 50 mg-30 g; 75 mg-20 g; 100 mg-20 g; 100 mg-10 g; 200 mg-8 g; 400 mg-6 g; 600 mg-4 g; 800 mg-3 g; 1 -2.5 g; 1 ,5-2 g; 5 mg-5 g; or 5 mg-50 g per day. Each dosage range represents a separate embodiment of the present invention.
[00135] In another embodiment, a composition of the present invention is administered at a dose that produces a desired effect in at least 10% of a population of treated patients. In another embodiment, the dose is that which produces the effect in at least 20% of treated patients. In other embodiments, the effect is produced in at least 30%, in at least 40%, in at least 50%, in at least 60%, in at least 70%, in at least 80%, or in at least 90% of the treated patients. In another embodiment, the effect is produced in over 90% of the patients. Each possibility represents a separate embodiment of the present invention.
[00136] In one embodiment, the subject of methods of the present invention is a mammal. In another embodiment, the subject is a human. In other embodiments, the subject is a rodent or a laboratory animal. In another embodiment, the subject is a male. In another embodiment, the subject is a female. In another embodiment, the subject is any other type of subject known in the art. Each possibility represents a separate embodiment of the present invention.
[00137] In one embodiment, the terms "administering" or "administration" refer to bringing a subject in contact with a compound of the present invention. In other embodiments, administration comprises swallowing or imbibing the composition of the present invention. In another embodiment, the step of administration utilizes a pharmaceutical composition, a nutritional supplement, or the like. Each possibility represents a separate embodiment of the present invention.
[00138] In one embodiment, administration is performed by the subject. In another embodiment, administration is performed by a care provider. In another embodiment, administration is performed by a third party Each type of administration represents a separate embodiment of the present invention.
[00139] In another embodiment, an additional therapeutic compound is administered to the subject as part of the method of the present invention. In another embodiment, the uridine or precursor, derivative or source thereof is the sole active ingredient in the composition. In another embodiment, the uridine or precursor, derivative or source thereof and choline or precursor, derivative or source thereof are the sole active ingredients in the composition. Each possibility represents a separate embodiment of the present invention. [00140] In one embodiment, the additional therapeutic compound is a drug that acts as a undine phosphorylase inhibitor; e.g. benzyl barbiturate or derivatives thereof. In another embodiment, the compound is a drug that increases uridine availability. In another embodiment, the compound is a uridine secretion-inhibiting compound, e.g. dilazep oi hexobendine, In another embodiment, the compound is a uridine renal transport competitors, e.g. L-uridine, L- 2',3'-dideoxyuridine, and D-2',3'-dideoxyuridine. In another embodiment, the compound is a drug which acts in synergy with uridine in generation of a phospholipid. In another embodiment, the compound is a compound which competes with uridine in kidney clearance, e.g. L-uridine, L-2',3'-dideoxyuridine, and D-2',3'-dideoxyuridine or mixtures thereof as disclosed in U.S. Pat. Nos. 5,723,449 and 5,567,689. In another embodiment, the compound is any other compound that is beneficial to a subject
[00141] In another embodiment, a method of the present invention causes one of the above effects by means of stimulating a P2Y receptor of a neural cell, neuron, or brain cell. In another embodiment, one of the above effects is caused partially as a result of stimulating a P2Y receptor of a neural cell or neuron. In another embodiment, one of the above effects is caused partially or fully by means of stimulating a P2Y receptor of another cell type. In another embodiment, one of the above effects is caused without stimulating a P2Y receptor. Each possibility represents a separate embodiment of the present invention.
[00142] In one embodiment, the stimulation of a P2Y receptor is mediated by uridine or a related compound in a composition of the present invention. In another embodiment, the uridine is converted to a second compound that stimulates a P2Y receptor in the cell. In one embodiment, the second compound is uridine-5'-triphosphate. In another embodiment, the second compound is any metabolic product known in the art of uridine or derivative or source thereof. Each compound represents a separate embodiment of the present invention. In other embodiments, the uridine or derivative or source thereof is converted into the second compound intracellularly or extracellularly In another embodiment, the uridine or derivative or source thereof is secreted from a cell after being converted into the second compound. In another embodiment, the uridine or derivative or source -thereof contacts a different cell after being- secreted from the cell in which it was converted to the second compound, and stimulates a P2Y receptor in the different cell. Each possibility represents a separate embodiment of the present invention. [00143] P2Y receptors are a family of receptors known to be involved in platelet activation and other biological functions. They are reviewed in Mahaul-Smith MP et al, Platelets. 2004 15 :131 -44, 2004
[00144] In one embodiment, the P2Y receptor of the present invention is a P2Y2 receptor. In another embodiment, the P2Y receptor is a P2Y4 receptor. In another embodiment, the P2Y receptor is a P2Y6 receptor. In another embodiment, the P2Y receptor is any other P2Y receptor known in the art. Each possibility represents a separate embodiment of the present invention.
[00145] In another embodiment, the P2Y receptor stimulates a second messenger In one embodiment, the second messenger is a G alpha protein. In another embodiment, the second messenger is a G alpha(q) protein. In another embodiment, the second messenger is cAMP. In another embodiment, the second messenger is any other second messenger known in the art.
Second messengers, and their associated signaling pathways, are well known in the art, and are described, for example, in Ferguson S, Pharm Rev 53: 1 -24, 2001 ; Huang E et al, Ann Rev Biochem 72: 609-642, 2003; and Blitterswijk W et al, Biochem J. 369: 199-21 1 , 2003. Each second messenger represents a separate embodiment of the present invention
[00146] In other embodiments, the second messenger stimulates a phospholipase C enzyme, modulates intracellular calcium levels, or increases protein kinase C activity. In one embodiment, one or more of the above pathways stimulates membrane production. In another embodiment, the second messenger modulates or stimulates another cellular pathway that stimulates membrane production. Each possibility represents a separate embodiment of the present invention.
[00147] In one embodiment, uridine or a related compound in a composition of the present invention stimulates a receptor other than a P2Y receptor
[00148] In another embodiment of the methods of the present invention, the uridine and/or choline is carried in the subjects' bloodstream to the subject's brain cell or neural cell. In another embodiment, the substance is carried by diffusion to the subject's brain cell or neural cell. In another embodiment, the substance is carried by active transport to the subject's brain cell or neural cell In another embodiment, the substance is administered to the subject in such a way that it directly contacts the subject's brain cell or neural cell. Each possibility represents a separate embodiment of the present invention [00149] In one embodiment, "pharmaceutical composition" refers to a therapeutically effective amount of the active ingredients, i.e the uridine and/or choline, together with a pharmaceutically acceptable carrier or diluent. "Therapeutically effective amount," in another embodiment, refers to that amount which provides a therapeutic effect for a given condition and administration regimen
[00150] In other embodiments, the pharmaceutical composition containing the uridine and/or choline is administered to a subject by any method known to a person skilled in the art, such as parenterally, paracancerally, transmucosally, transdermal Iy, intramuscularly, intravenously, intradermally, subcutaneously, intraperitonealy, intraventricular^, intracranially, intravaginally or intratumoraUy.
[00151 ] In another embodiment, the pharmaceutical compositions are administered oially, and thus is formulated in a form suitable for oral administration, i.e. as a solid or a liquid preparation. Suitable solid oral formulations include, for example, tablets, capsules, pills, granules, pellets and the like. Suitable liquid oral formulations include solutions, suspensions, dispersions, emulsions, oils and the like In one embodiment of the present invention, the composition containing the uridine and choline is formulated in a capsule. In accordance with this embodiment, the compositions of the present invention comprises a hard gelating capsule, in addition to the active compounds and the inert carrier or diluent.
[00152] In another embodiment, the pharmaceutical compositions are administered by intravenous, intraarterial, or intramuscular injection of a liquid preparation, Suitable liquid formulations include solutions, suspensions, dispersions, emulsions, oils and the like. In one embodiment, the pharmaceutical compositions are administered intravenously, and are thus formulated in a form suitable for intravenous administration. In another embodiment, the pharmaceutical compositions are administered intraarterial Iy, and are thus formulated in a form suitable for intraarterial administration In another embodiment, the pharmaceutical compositions are administered intramuscularly, and are thus formulated in a form suitable for intramuscular administration.
[00153] Further, in another embodiment, the pharmaceutical compositions are administered as a suppository, for example a rectal suppository or a urethral suppository. In another embodiment, the pharmaceutical compositions are administered by subcutaneous implantation of a pellet. In another embodiment, the pellet provides for controlled release of uridine and/or choline over a period of time. [00154] Pharmaceutically acceptable carriers or diluents are well known to those skilled in the art. The carrier or diluent is, in one embodiment, a solid carrier or diluent for solid formulations, a liquid carrier or diluent for liquid formulations, or mixtures thereof.
[00155] Solid caπiers/diluents include, in other embodiments, a gum, a starch (e.g. corn starch, pregeletanized starch), a sugar (e.g., lactose, mannitol, sucrose, dextrose), a cellulosic material (e.g. microcrystalline cellulose), an acrylate (e.g. polymethylacrylate), calcium carbonate, magnesium oxide, talc, or mixtures thereof.
[00156] For liquid formulations, pharmaceutically acceptable carriers are, in other embodiments, aqueous oi non-aqueous solutions, suspensions, emulsions or oils. Non-aqueous solvents include propylene glycol, polyethylene glycol, and injectable organic esters such as ethyl oleate. Aqueous earners include water, alcoholic/aqueous solutions, emulsions oi suspensions, including saline and buffered media Examples of oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, mineral oil, olive oil, sunflower oil, and fish-liver oil.
[00157] In another embodiment, the compositions further comprise binders (e.g. acacia, cornstarch, gelatin, carbomer, ethyl cellulose, guar gum, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, povidone), disintegrating s (e g cornstarch, potato starch, alginic acid, silicon dioxide, croscarmelose sodium, crospovidone, guar gum, sodium starch glycolate), buffers (e.g., Tris-HCL, acetate, phosphate) of various pH and ionic strength, additives such as albumin or gelatin to prevent absorption to surfaces, detergents (e.g., Tween 20, Tween 80, PIuronic F68, bile acid salts), protease inhibitors, surfactants (e g sodium lauryl sulfate), permeation enhancers, solubilizers (e.g., glycerol, polyethylene glycerol), anti-oxidants (e.g , ascorbic acid, sodium metabisulfite, butylated hydroxyanisole), stabilizers (e.g. hydroxypropyl cellulose, hyroxypropylmethyl cellulose), viscosity increasing s(e.g. carbomer, colloidal silicon dioxide, ethyl cellulose, guar gum), sweetners (e.g. aspartame, citric acid), preservatives (e g , Thimerosal, benzyl alcohol, parabens), lubricants (e.g stearic acid, magnesium stearate, polyethylene glycol, sodium lauryl sulfate), flow-aids (e g colloidal silicon dioxide), plasticizers (e g. diethyl phthalate, triethyl citrate), emulsifϊers (e.g. carbomer, hydroxypropyl cellulose, sodium lauryl sulfate), polymer coatings (e g., poloxamers or poloxamines), coating and film forming s (e.g. ethyl cellulose, acrylates, polymethacrylales) and/or adjuvants [00158] In another embodiment, the pharmaceutical compositions provided herein are controlled release compositions, i.e. compositions in which the uridine and/or choline is released over a period of time after administration. Controlled or sustained release compositions include formulation in lipophilic depots (e.g. fatty acids, waxes, oils). In anothei embodiment, the composition is an immediate release composition, i.e. a composition in which all of the uridine and/or choline is released immediately after administration
[00159] In another embodiment, the pharmaceutical composition is delivered in a controlled release system. For example, the composition is administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration. In one embodiment, a pump is used (see Langer, supra; Sefton, CRC Crit Ref Biomed Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J Med. 321 :574 (1989). In another embodiment, polymeric materials are used. In another embodiment, a controlled release system is placed in proximity to the theiapeutic target, i.e., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol 2, pp. 1 15-138 (1984) Other controlled release systems are discussed in the review by Langer (Science 249.1527-1533 (1990).
[00160] The preparation of pharmaceutical compositions which contain an active component is well understood in the art, foi example by mixing, granulating, or tablet-forming processes. The active therapeutic ingredient is often mixed with cxcipients which are pharmaceutically acceptable and compatible with the active ingredient. Foi oral administration, the uridine and/or choline or their physiologically tolerated derivatives such as salts, esters, N- oxides, and the like are mixed with additives customary for this purpose, such as vehicles, stabilizers, or inert diluents, and converted by customary methods into suitable forms for administration, such as tablets, coated tablets, hard or soft gelatin capsules, aqueous, alcoholic or oily solutions. For parenteral administration, the uridine and/or choline or their physiologically tolerated derivatives such as salts, esters, N-oxides, and the like are converted into a solution, suspension, or emulsion, if desired with the substances customary and suitable for this purpose, for example, solubilizers or-other.
[00161 ] An active component can be foimulated into the composition as neutralized pharmaceutically acceptable salt forms Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide or antibody molecule), which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed from the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like
[00162] For use in medicine, the salts of the uridine and/or choline are pharmaceutically acceptable salts. Other salts are, in one embodiment, useful in the preparation of the compounds according to the invention or of their pharmaceutically acceptable salts. Suitable pharmaceutically acceptable salts of the compounds of this invention include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulphuric acid, methanesulphonic acid, fumaric acid, maleic acid, succinic acid, acetic acid, benzoic: acid, oxalic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
EXPERIMENTALDETAILSSECTION EXAMPLE1
Measurement Of Cvtidine By HPLC Without Interference From Tyrosine
MATERIALS AND METHODS
Sample preparation
[00163] 1 -milliliter (mL) samples of heparinized plasma were spiked with 1 μg fluoro- uiidine for use as an internal standaid. then deproteinized by adding methanol (5 mL). Samples were centrifuged, lyophilized, reconstituted in 5 mL of 0.25 N ammonium acetate (pH 8.8), then immediately purified over boronate affinity columns,
Boronate affinity columns
[00164] All steps were performed at 4° C Boronate affinity columns (Affigel-601 , Bio-
Rad) were primed with two 5-mL ammonium acetate washes, samples were applied, and columns were washed again with ammonium acetate, after which the nucleosides were eluted with 0.1 N formic acid (7 mL). Eluates were lyophilized, then reconstituted in 100 μL water for HPLC analysis Boronate affinity columns bind many biological molecules, including the nucleotide bases adenosine, cytidine, guanosine, thymidine, and uridine.
HPLC [00165] HPLC analysis was performed using a Beckman System Gold apparatus
(Beckman Instruments) equipped with a Rainin Dynamax Microsorb Cl 8 column (3 μm packing; 4.6 X 100 mm) at room temperature. The standard HPLC method is described in Lopez-Coviella et al, (J. Neurochem 65: 889-894, 1995). For modified HPLC, an isocratic elution buffer was used containing 0.004 N potassium phosphate buffer (pH 5 8) and 0.1% methanol instead of formic acid, flowing at 1 mL/min and heated to 35°.
RESULTS
[00166] A standard HPLC method for measuring nucleosides yields separate peaks for uridine and cytidine; however, a coincidence of the cytidine and tyrosine peaks precludes accurate measurement of cytidine levels, as shown for human plasma samples (Figure 1 ). Tyrosine is present in many biological fluids, e.g., plasma or cerebrospinal fluid (CSF) In the present Example, a modified HPLC method was used which distinguished cytidine and tyrosine peaks, permitting accurate measurement of cytidine levels (Figure 2).
EXAMPLE 2
Oral administration of UMP increases plasma uridine levels in humans
MATERIALS AND EXPERIMENTAL METHODS Study design
[00167] Eight healthy subjects (5 male, 3 female, 27-67 years old) were instructed to fast overnight and given sequentially increasing doses (500, 1000, and 2000 mg) of disodium UMP (Numico, Wageningen, NL) at 7 - 8 AM on each of three days, sepaiated by at least a three-day washout period. All subjects were given lunch. Blood samples were drawn over an eight-hour period into heparinized tubes Plasma was treated with methanol to precipitate protein, extracted with chloioform, and an aliquot of the aqueous layer lyophilized, dissolved in water, and assayed by HPLC with UV detection.
Statistical analyses
[00168] Statistical analyses were carried out with SPSS 12.0. Data were represented as mean ± SEM. Unpaired Student's t test, one-way analysis of variance (ANOVA), ANOVA with repeated measures, two-way ANOVA were used to assess the statistical effects, as described in detail in the context Tukey's HSD post hoc analyses were conducted when appropriate The significance level was set atp O.05.
RESULTS
[00169J Subjects were administered 500, 1000, or 2000 mg UMP orally, and blood uridine levels were measured at baseline and 1, 2, 4 and 8 hours (hr) following dosing Plasma uridine levels were assayed as described in Example 1. Plasma uridine levels increased in response to oral UMP in a dose-dependent fashion, then returned to baseline levels within 8 hr (Figure 3).
EXAMPLE 3
Oral administration of uridine or UMP increases plasma uridine levels in gerbils
MATERIALS AND EXPERIMENTAL METHODS Experimental design
[00170] Groups of eight to nine male gerbils (60-80 g) were fasted overnight, administered (a) uridine (Sigma, St Louis, MO; 250 mg/kg body weight) (Figure 4) or disodium UMP (1 mmol/kg body weight, a dose equivalent to 250 mg/kg uridine by gavage) (Figure 5) and sacrificed by decapitation under Telazol anesthesia one hour later For Figure 6, gerbils were fed chow (Harlan Teklad, Madison, WI) ad lib containing either 0. 0 1 , 0.5 or 2.5% UMP by weight for 4 weeks, fasted overnight, then sacrificed one hour after consumption of a last meal of the same composition. Blood collected from the neck was collected into tubes containing EDTA and was treated as described above for Example 2.
RESULTS
[00171] To ascertain whether oral administration of uridine can raise plasma uridine levels, gerbils were fed by gavage 250 mg/kg cytidine or uridine 60 minutes (min) later, plasma uridine levels were assessed by the method described in Example 1. Both dietary cytidine and uridine increased plasma uridine levels by a statistically significantly margin relative to a control group that was fed chow not containing cytidine or uridine, both dietary uridine resulted in plasma uridine levels approximately 3-fold higher than dietary cytidine (Figure 4)
[00172] In a separate experiment to assess the time course of the increase in plasma uridine levels, gerbils were administered either water or 1 mϋlimole (mmol) UMP per kilogram (kg) body weight, were sacrificed at various time points in the following 60 min, and plasma uridine levels were assessed Plasma uridine levels increased within 10 min of administration, reaching peak levels by 30 min (Figure 5).
[00173] In another experiment, gerbils were fed either a control diet or a diet containing 0.1%, 0.5%, or 2 5% LIMP. One hour later, plasma uridine levels were assessed As depicted in Figure 6, plasma uridine levels increased in response to dietary UMP in a dose-dependent manner. These results indicate that orally administered uridine is absorbed into the bloodstream
EXAMPLE 4 Oral administration of uridine or UMP increases brain uridine levels in gerbils
MATERIALS AND METHODS Gerbil brain tissue preparation
[00174] Brains were quickly removed from the skull after decapitation, frozen on dry ice, homogenized in 80% methanol, centrifuged, lyophilized and analyzed as described for Example 3
RESULTS
[00175] To ascertain whether oral administration of uridine can raise brain uridine levels, brains of the gerbils from the first experiment in Example 3 were homogenized, and the uridine levels were assayed. Oral administration of cytidine resulted in a two-fold increase in brain uridine levels, and oral administration of uridine resulted in a greater than a three-fold increase in brain uridine levels, relative to the control animals (Figure 7). All differences between groups were statistically significant
[00176] In order to assess the time course of the increase in plasma uridine levels, brain uridine levels were assessed in the gerbils from the second experiment of Example 3 Brain uridine levels increased within 10 min of uridine administration, reaching peak levels within 30 mhvsimilar to the results observed with plasma uridine levels (Figure 8). These results indicate that orally administered uridine is efficiently transported into the brain
EXAMPLE 5
Uridine is Readily Converted to Cvtidine in the Brain [00177] In a separate experiment, gerbils were orally administered 250 mg/kg body weight uridine, and 60 min later plasma and brain levels of cytidine and uridine were assessed. The fold-increases relative to control animals was calculated and are depicted in Figure 9A (plasma) and Figure 9B (brain). In each case, the fold-increase of cytidine was normalized to the fold increase of uridine, which was arbitrarily set as 100%. These results indicate that (a) uridine in the bloodstream is transported into the brain and (b) uridine is metabolically processed differently in the brain than in plasma; specifically, it is more efficiently converted to cytidine than in plasma.
EXAMPLE 6
Uridine Increases Levels of the Phospholipid Precursor CDP-Choline in the Brain and in a Neural Cell Line
METHODS
Experimental design
[00178] Data was pooled from three experiments, with group sizes ranging from 5 to 16 animals. Male gerbils (60 - 80 g) were given UMP (1 mmole/kg body weight) by gavage and sacrificed at the indicated times. After brain homogenization, protein precipitation, and lyophilization as described for Example 4, samples were analyzed by HPLC-UV.
Assessment of CDP-choline levels
[00179] Brain tissue or cells was dissolved in methanol/chloroform (1 :2 vol/vol), centrifuged, and the aqueous phase was dried under vacuum, resuspended in 100-200 μL water and separated by HPLC on an ion-exchange column (Alltech Hypersil APS-2, 5 μM, 250 x 4.6 mm). CDP-choline was eluted with a linear gradient of NaH2PO4 buffers A (1.75 mM NaH2PO4, pH 2.9) and B (500 mM, pH 4.5), which allowed resolution of CDP-choline from closely co-eluting substances such as UMP over 40 min. The retention time for CDP-choline was_ 9.5 min. Individual nucleotide peaks were detected by UV absorption at 380 nrn, and were identified by comparison with the positions of authentic standards, as well as by the addition of nucleotide standards to selected samples.
PCl 2 cells [00180] PC12 cells were maintained in Minimal Essential Medium (MEM; Invitrogen,
Carlsbad, CA) supplemented with 10% fetal bovine serum (FBS) at 37 0C. Experimental incubations were for 2 or 4 days in medium containing 50 ng/ml mouse 2.5 S (2 5 subunit) NGF and 1 % FBS, with or without test compounds NGF and FBS were obtained from Invitrogen
RESULTS
[00181 ] In order to assess the effect of orally administered uridine on levels of phospholipid precursors in the brain, brains of the gerbils from the second experiment of Example 3 were assayed for levels of CDP-choline, a key intermediate in phospholipid biosynthesis via the Kennedy pathway. Levels of CDP-choline rose significantly in a linear fashion (regression analysis, r = 0.98, p < 0.02) for 30 min after administration of UMP (Figure 10).
[00182] To directly demonstrate conversion of uridine to CDP-choline in neural cells, PC
12 cells, a cell line capable of differentiation into neural cells, were treated with uridine, and intracellular levels of CDP-choline were measured. Uridine treatment resulted in a statistically significant increase in CDP-choline levels after 50 minutes (Figure 1 1 ). These results show that, after transport to the brain, uridine is converted to phospholipid precursors such as CDP- choline, perhaps via the intermediate CTP, and therefore augments cognitive function by increasing synthesis of phospholipid precursors in brain cells.
EXAMPLE 7
Oral Administration of UMP Increases Neurotransmitter Release in Brains of Aged Rats
METHODS
Animals and dietary UMP supplementation
[00183] Male middle aged Fischer 344 rats, 22-24 months old at the time of doing microanalysis, were obtained from National Institute on Aging "(Harlan Sprague-Dawley, Indianapolis, IN). Rats were housed individually under standard husbandry conditions and exposed to 12 hr light/ dark cycle with food and water provided ad libitum Each rat consumed approximately 500 mg/kg/day of UMP-2Na (LD50 by ip of uridine is about 4.3 g/Kg). [00184] Rats were acclimated to the animal facility for more than 7 days before fed a control laboratory diet (Teklad Global 16% protein rodent diet, TD.00217, Harlan Teldad, Madison, WI), or this diet fortified with UMP«2Na+ (2.5%, TD.03398, UMP«2Na+; Numico Research, the Netherlands) for 6 weeks.
[00185] Rats were not fed with the research diet until at least 7 days later after their arrival. They were weighed at the time of beginning feeding (t=0), as well as 1 , 2, 4, 6 weeks later. Al time 0, rats were randomly assigned into two gioups. There were no significant differences of body weight between groups (FIJ I = 3.03,^7 > 0-05); average weight was 455 ± 5 (N = 13 rats). Repeated measures with weeks as within-subjects factor showed feeding time (0, 1 , 2, 4, 6 weeks) significantly changed body weight (^4,44 = 2.65, p < 0 05), while neither UMP- diet (vs. control) nor UMP*time interaction affected body weight (Fu ] = 0.01 , F^M = 1 -25, respectively; all p > 0 05)
[00186] The experiment described in this Example was performed twice, each time with
7 control rats and 9 rats administered the UMP diet
Chemicals and solutions
[00187] Dopamine (DA), dihydroxyphenylacetic acid (DOPAC), homovanillic acid
(HVA), serotonin (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), and 3,4-dihydroxybenzoic acid (DHBA; internal standard) were purchased from Sigma (St Louis, MO) and were dissolved in HCIO4 (0.1 M) to make 1 mM stock solutions, and aliquots were kept at -8O0C. Ketamine hydrochloride (100 mg/ml) was purchased from Fort Dodge Animal Health (Fort Dorge, IA). Xylazine (20 mg/ml) originated from Phoenix Scientific, Inc. (St. Joseph, MO).
[00188] Ringer solution consisted of NaCl 147, KCl 2.7, CaCl2 1.2 and MgCl2 0 85 mM.
For high potassium solution, KCl was increased to 80 mM, with NaCl decreased to 69 7 mM to maintain osmolarity. All solutions were made from doubly distilled deionized water and filtered by Steriflip® (Millipore, Bedford, MA)
In vivo microdialysis
[00189] Rats were anesthetized with a mixture of ketamine and xylazine (80 and 10 mg/Kg of body weight, respectively, intraperitoneally), and were placed in a Kopf stereotaxic frame All surgical instruments were sterilized by a hot bead dry sterilizer or 70% ethanol. A small hole was drilled into the skull by a 2-mm trephine bone drill. CMA/1 1 14/04 Cupr piobe (O.D. 0.24 mm, 4 mm membrane, 6,000 Da, CMA microdialysis, Sweden) was implanted into the right striatum (AP = +0.5, ML = -3.0 from Bregma, DV = -7.3 mm from Dura, as described in Paxinos G et al, The Rat Brain in Stereotaxic Coordinates, 2nd ed., Academic Press, San Diego) with incisor bar set at -5.0 mm. Probes were secured permanently in position using dental cement and three anchor screws to the skull. After surgery, rats were injected intraperitoneally with saline (5 ml/kg) and kept on a heating pad maintaining body temperature at 37°C until awaking.
[00190] The freely moving rat was perfused in a circular bowl on a rotating platform obviating the need for a liquid swivel (see Wang L el al, Neurochem Int 42: 465-70, 2003), and was habituated to the environment on the first day after surgery. Experiments were performed approximately 48 Iir after the surgery, and were carried out between 10:00 am to 4:00 pm. Ringer's solution was perfused continuously using Fluorinatedelhylenepropylene (FEP) Resin tubing and a gas-tight syringe (Exmire type I, CMA), at a constant rate of 1 5 μl/min by a microinfusion pump (CMA/100). Dialysates were collected at 15-min intervals. 5 μl of antioxidant mixture, consisting of 0.2 M HClO4 and 0.1 mM EDTA, was added to the sampling vial prior to collection to protect dopamine and its metabolites. The samples within the first 60 min were discarded from analysis. Subsequently, 3 consecutive sessions of samples were collected. Except for the last session (1 5 hrs, 6 samples), the others were collected for 1 hr (4 samples) The order was as follows: session 1 (aCSF), 2 (High K+), 3 (aCSF). All samples were collected on crushed ice, instantly frozen and kept at -80 0C until HPLC analysis.
Brain dissection for the proteins and monoamines
[00191] After microdialysis experiments, rats were anesthetized with ketamine and xylazine (80 and 10 mg/Kg, i p ). A black ink was pushed through the probe to stain the tissue around the probe Rats were decapitated with a guillotine. Brains were quickly dissected on a chilled dissection board. The left striatum was snap-frozen in an Eppendorf tube placed in liquid nitrogen for future protein assays. The right striatum was further dissected, and the position of probe was determined by visual observation. Data were not included if probe was found not within the striatum
[00192] An additional group of rats (20 months old; n = 6 for both control and UMP) were fed for 6 weeks No microdialysis was caπied out in these rats. Striata (both left and right) were collected as above to determine tissue levels of dopamine and its metabolites.
Extraction of tissue dopamine samples [00193] The striatum were weighed and homogenized in an Eppendorf tube on ice for 1 min with 1 ml of H2O containing 0 1 M HClO4 and 1 μM EDTA. After vortexing for 10 seconds, an aliquot was used for Bicinchoninic Acid (Sigma, St Louis, MO) protein assay. The homogenates were then filtered with Ultrafiee-MC centrifugal filter units (Millipore, 14,000 rpm/15 min/4 0C) A 1 : 10 dilution was made before the aqueous layer was subjected to HPLC DHBA was added to the samples prior to homogenization as the internal standard. Concentrations of dopamine and its metabolites were determined by HPLC, and values from the three repeated measures were averaged and normalized to the amount of protein per sample.
Analysis of dopamine and metabolites [00194] DA and metabolites in dialysates and tissue samples were determined using an
ESA Coulochem 11 5100A detector (E i = -175 mV; E2 = +325 mV, Eguard = 350 mV) with an ESA Microdialysis Cell (model 5014B, ESA, North Chelmsford, MA) The mobile phase (MD- TM, ESA) consisted of 75 mM NaH2PO4, 1 7mM 1 -octanesulfonic acid, 100 μl/L Triethylamine, 25 μM EDTA, 10% acetonitrile, pH 3.0. The flow rate was 0.4 mL/min. The column (ESA MD 150, 3X 150 mm, 3 μm, 120 A) was kept in a 40 0C column oven Samples were injected to HPLC by an Λlltech 580 autosamplei (Alltech, Deerfield, IL) and maintained to 4°C with a cooling tray during analysis Data were captured by Alltech AllChiom " data system, and analyzed with AllCluom plus "' software A timeline program, which could change the detection gain during sample separation and detection, was used to make it possible to get low DA and high metabolites concentration data in dialysate through one injection.
Data analysis
[00195] Data were represented according to sampling time of six to nine measurements each point (means ± standard error of measurement [S.E M.]) Basal values of DA and major metabolites were determined based on the averages of the first four consecutive samples prior to K"1 stimulation (mean value in the dialysate was 10 2 ± 0.4 nM, n = 22), which was assigned a value of 100%. Statistics were performed using two-way ANOVA (Treatmentxtime) with Turkey's HSD post hoc test One-way ANOVA was used to compare the differences among the tHree" groups" in each time point. A p value" of > 0 05~was~ usedlo assess"statislical significance". Basal levels of dopamine were homogeneous between the two replicated experiments and were therefore pooled into the corresponding groups (TM ^o = 3.99, p > 0 05). Basal DA levels in the dialysates were stable after 1 hr equilibration, in the four consecutive samples prior to K+ stimulation (^3,57 = 0.15, p > 0.05; one-way ANOVA with repeated measures -using sampling time (0, 15, 30, 45 min) as within-subjects factor).
[00196] Similar to basal DA levels, basal levels of DOPAC and HVA in the dialysates were 612 ± 14 and 369 ± 7 nM (n = 22 rats), and were stable (^57 = L06, F3-57 = 0.84, respectively; in each case, p > 0.05). There were no effects of UMP treatment on the basal DOPAC and HVA levels (Control vs. UMP-I week vs. UMP-6 weeks; F2-19 = 0 27, F2, 19 = 0.03, respectively; in each case, /7 > 0.05).
RESULTS [00197] In order to assess the effect of orally administered uridine metabolites on neurotransmitter release in the brain, aged rats maintained in a restricted environment consumed for 1 or 6 weeks either a control diet 01 a diet supplemented with 2.5% UMP UMP supplementation did not affect basal DA levels in the dialysate among treatment groups (control vs. UMP- 1 week vs. UMP- 6 weeks; F2, 19 = 0.98) DA concentration in the dialysate was 10.2 ± 0 4 nM (n = 22 rats)
[00198] The effect of dietary UMP supplementation on K"1 -evoked striatal DA release
(following perfusion with the high-K+ solution) is depicted in Figure 12A. A statistically significant difference (F2-2Oe = 3.36) was found in DA levels in the dialysates among the control, UMP- 1 week, and UMP- 6 weeks treatment groups. Post hoc multiple comparisons revealed a significant difference between control and UMP-6 weeks' groups. Data were further divided into three sections (before, K+-evoked and after), which also revealed a significant enhancement of K+-evoked DA release between control and UMP-6 weeks' groups, from 283 ± 9% to 341 ± 21 % (Figure 12B). The UMP-I week group also exhibited increased DA release (316 ± 15%) relative to the control group; however, this increase was not significant.
[00199] Next, the effect of dietary UMP supplementation on the DA metabolites 3,4- dihydroxyphenylacetic acid (DOPAC) and homovanilic acid (HVA) in striatal dialysate was assessed K+-depolarization, significantly deceased DOPAC (Figure 13A) and HVA (Figure 13B) to 65 ± 4% and 51 ± 4% compared to baseline levels in all groups (F2,95 = 51 90, F2^5 = 92.74, respectively; all /? < 0.01)- There were no differences in K+-decreased DOPAC and HVA levels among treatment groups (F2-2Gf1 = 1.01, F2.266 = 1 20, respectively). Changing the solution from high K+ back to normal Ringer's solution at 105 min increased both DOPAC and HVA levels in the dialysate, with maximum levels attained al 30 min after changing (DOPAC, 169 ± 9%; HVA, 149 ± 5%). However, no significant differences were found among the three groups
[00200] In addition, dietary UMP was shown to increase the basal release of the neurotransmitter acetylcholine fiom neurons in the corpus striatum (Figure 14).
[00201] These results show that (a) oially administered uridine improves neurotransmitter release in the brain; (b) uridine-mediated augmentation of brain function is a multi-species phenomenon, not limited to gerbils; and (c) augmentation of brain function by uridine occurs biologically relevant animal model for age-impaired cognitive dysfunction.
EXAMPLE 8
Oral Administration of UTP Increases Levels of NF-70 and NF-M in Brains of Aged Rats
METHODS
Data analysis
[00202] Data were represented according to UMP treatment of six to sixteen measurements each group (means ± S-E-M ). One-way ANOVA with Turkey's HSD post hoc tests were used to compare the difference among the treatments the Newman-Keuls multiple range test was used for the data in Figure 16.
Western blotting
[00203] Striatal tissues were placed in Eppendorf tubes containing 200 μl lysis buffer (60 mM Tris-HCl, 4% SDS, 20% glycerol, 1 mM dithiothreitol, 1 mM AEBSF, 8 μM aprotinin, 500 μM bestatin, 15 μM E64, 200 μM leupeptin, 10 μM pepstatin A). The samples were sonicated, boiled (10 min), and centrifuged (14,000 g for 1 min at room temperature). The supernatant fluid was transferred to a clean tube, and total protein content was determined using the Bicinchoninic Acid assay (Sigma, St Louis, MO).
[00204] Equal amounts, of protein (40 μg protein/lane) were loaded /or sodium jdodecyl sulfate-polyacrylamide gel electrophoresis (4-15% SDS PAGE; Bio-Rad, Hercules, CA) Prior to gel electrophoresis, bromphenol blue solution (0.07%) was added to each sample Proteins were separated, transferred onto polyvinylidene difluoride (PVDF) membranes (Immobilon-P, Millipore), and blocked with 5% bovine serum albumin (Tris-buffered saline/0.15% Tween 20) for 1 h. After 3 10 min rinses in Tris-buffered saline (TBST), blots were incubated in TBST with various antibodies against the proteins of interest, including NF-70, NF-M (1 : 2000, 1 : 5000, respectively; Calbiochem, La .lolla, CA) at 4 °C overnight on an orbital shaker. Protein- antibody complexes were detected and visualized using the ECL system (Amersham, Piscataway, NJ) and Kodak X-AR film, respectively, as suggested by the manufacturer. Films were digitized using a Supervista S-12 scanner with a transparency adapter (UMAX Technologies, Freemont, CA). Analysis was performed using the public domain NIH Image program (NIH V.I 61).
RESULTS [00205] In order to assess whether incieasing uiidine levels can augment the production of new membrane in the brain, levels of neurofilament-70 (NF-70) and neurofilament-M (NF- M), biomarkers of neurite outgrowth, were assessed in the biains of the rats fiom the experiment described in Example 7 As shown in Figure 15, UMP dietary supplementation for 6 weeks significantly increased the levels of NF-70 (Figure 15A) and NF-M (Figure 15B), to 182 ± 25% (F2.31 = 6 01 , p < 0.05) and 221 ± 34% (F2,21 = 8 86, p < 0.01 ) of control values, respectively. Consumption of a UMP diet for 1 week did not increase the levels of these two proteins compared to control gioup in a statistically significant manner. Levels of NF-70 and NF-M in striatum increased to 204 ± 36% and 221 ± 34% of control values, respectively.
EXAMPLE 9
Oral Administration of Uridine or UTP Increases Neurite Outgrowth and Branching and
Levels of NF-70 and NF-M in PC 12 cells
METHODS Data analysis
[00206] Data are presented as mean +/- S.E.M. Analysis of variance (ANOVA) was used to determine differences between groups (significance level, p<0.05). When differences were detected, means were separated using the Newman-Keuls multiple range test
Neurite outgrowth studies [00207] PC 12 cells were sparsely plated on collagen-coated 60 mm culture dishes in
MEM containing 1% fetal bovine serum. Experimental groups were as follows: undine, uridine triphosphate, cytidine, reactive blue 2, suramin and PPADS (Sigma, St. Louis, MO). All treatments were performed 24 h after plating. At the end of the treatment period, images were obtained with a phase-contrast Zeiss Axioplan 2 microscope, using OpenLab software. Six digital images were captured for each dish, for a total of 18 to 24 images per treatment group. Approximately 300 cells were quantified for each treatment group for each experiment Experiments were performed in triplicate. Quantification of neurites, including neuiite branching and neurite length, was performed by one more researchers blinded to experimental gioups. Neurite length was measured using the public domain NIH software "Image J." Processes longer than the diameter of the cell body were counted as neurites. Only process- bearing cells were analyzed.
Detection of intracellular UTP and CTP
[00208] Levels of intracellular UTP and CTP were analyzed by HPLC as described for
Example 6, except that 5 mM NaH2PO,!, pH 2.65 was used as buffer A
RESULTS
[00209] The effect of uridine treatment (10 - 200 μM) on NGF-induced neurite outgrowth was next tested In the absence of NGF, PC 12 cells did not sprout neurites (fewer than 1 Vo). Uridine treatment (50 μiM, 2 or 4 days) in the absence of NGF did not result in the production of neurites. In the presence of NGF, uridine (50 - 200 μM) significantly (p < 0.01 oi 0 001 ) enhanced the number of neurites per cell after 4 days of treatment (Figure 16A-C), whereas 2-day treatment or lower uridine concentrations (10, 25 μM) had no effect. Treatment of the NGF-exposed cells with cytidine also had no effect on neurite outgrowth.
[00210] Since uridine increased the number of neurites per cell, the effect of uridine on neurite branching and length in the presence of NGF was also assessed. After 4 days of tieatment with uridine (50 μM) and NGF, the numbers of neurite branch points per cell were significantly (p < 0.01) increased, compared with those in cells treated with only NGF (Figure 16D). Uridine did not significantly affect average neurite length in NGF-differentiated cells
[002Ii] Neurofilament proteins are highly enriched within neurites;~therefore, an~ increase in neurile number should be associated with increased expression of neurofilament proteins NF-70 (70 kD) and NF-M (145 IcD) levels following 4-day treatment of PC 12 cells with NGF alone, or NGF plus uridine (50 μM) were thus measured (Figure 16E). Both NF-70 and NF-M expression significantly (p < 0.01, p < 0.001 , respectively) increased following uridine treatment, compared Io cells treated only with NGF In the absence of NGF, uridine treatment had no effect on levels of either neurofilament protein. Thus, uridine augments neurite outgrowth in PC 12 cells
[00212] In the absence of NGF, the addition of exogenous uridine increases intracellular LJTP and CDP-choline levels in PC 12 cells (Example 6). To determine whether uridine affects
UTP or CTP levels in the presence of NGF, levels of UTP and CTP were measured in PC 12 cells for 2 days with NGF, treated with no nucleotide, (control), uridine, cytidine or UTP, in the presence of NGF Uridine (50 μM) significantly (p < 0.05) increased both UTP and CTP levels
(Figure 17 A-B, respectively) compared to cells receiving only NGF treatment UTP (100 μM) or cytidine (50 μM) did not significantly affect the intracellular levels of either nucleotide,
[00213] In order to ascertain whether UTP may mediate the effect of uridine on neurile outgrowth, PCl 2 cells were treated with NGF and various doses of UTP. After 4 days of treatment, UTP (10 and 50 μM) significantly (p < 0.01) enhanced neurite outgrowth, compared to that in cells treated only with NGF. Thus, either uridine or UTP augments neurite outgrowth.
[00214] In conclusion, uridine or UTP dietary supplementation increased the levels of two major neurofilament proteins in rat brain, and was directly shown to induce neurite outgiowth in PC 12 cells.
EXAMPLE 10
NGF-differentiated PC 12 cells express pyrimidine-sensitive P2Y2, P2Y4 and P2Y6 receptors
METHODS
Detection of P2Y receptors
[00215] Western blots were performed as described for Example 8, using rabbit anti-
P2Y2, anti-P2Y4 (both from Calbiochem); or rabbit anti-P2Y6 (Novus Biologicals, Littleton. " CO).
Immunocytochemistry
[00216] PC 12 cells were treated as described above, except they were grown on 12mm glass cover slips (A Daigger & Co , Vernon Hills, IL) coated with collagen. Proteins were visualized using immunofluorescence. Briefly, the cells were fixed with 4% paraformaldehyde, permeabilized with 0.25% Triton X-I OO, blocked in 10% normal goat serum, and incubated overnight in the appropriate antibodies (mouse anti-NF-70, and either rabbit anti-P2Y2, rabbit anti-P2Y4 or rabbit anti-P2Y6). For P2Y2 and P2Y4 visualization, control cultures weie incubated with primary antibody plus a control antigen in order to ensure that the immuno- staining would be specific. Control antigen was not available for the P2Y6 receptor Cells were then incubated in fluorochrome-conjugated secondary antibodies for 1 hour (goat anti-rabbit ALEXA 488 and goat anti-mouse ALEXA 568; Molecular Probes, Eugene, OR) and mounted on glass slides with mounting media with or without DAPI (Vector Laboratories, Burlingame, CA). Control antigens provided with the primary antibodies were used to ensure that immuno- staining was specific Digital images were obtained on a Zeiss (Oberkochen. Germany) Axioplan microscope with OpenLab software, using a Zeiss Plan-Neofluor 4Ox oil-immersion objective.
RESULTS [00217] UTP is an agonist of the pyrimidine-activated class of P2Y receptors, namely
P2Y2, P2Y4 and P2Y6 receptors. To determine whether these receptors participate in the mechanism by which extracellular UTP affects neuritc outgrowth, it was first determined whether the receptors are expressed in PC 12 cells, and whether exposure to NGF alters their expression, PC 12 cells were treated for 0 - 7 days with NGF and levels of the receptors measured. After 3 days of NGF treatment, expression of the P2Y2 receptor reached maximal levels, which were significantly (p < 0.001) higher than those seen at less than 3 days of NGF treatment (Figure 19A). To visualize the expression and localization of the P2Y2, as well as the P2Y4 and P2Y6, receptors, cells were grown in the presence or absence of NGF for 4 days and then immuno-stained them for the neuritic marker NF-70, and for P2Y2, P2Y4, or P2Y6 (Figure 19B, left to right, respectively). All three receptors were highly expressed in NGF- differentiated PC 12 cells In addition, P2Y2 co-localized with the neuronal marker MAP-2 (Figure 20) In the absence of NGF, receptor protein expression was undetectable by immuno- staining Moreover, the presence of uridine did not affect the expression of the receptors compared "with the quantities present in cells exposed to NGF alone. Thus, the P2Y2, P2Y4 and" P2Y6 receptors are present in neural cells, but not in their precursors.
EXAMPLE Il Antagonism of P2Y receptors inhibits the effect of uridine on NGF-induccd ncurite outgrowth
[00218] To ascertain whether signaling by P2Y receptors mediate induction of neurite outgrowth by uridine, PC 12 cells were incubated for 4 days with NGF, uridine (100 μM) and the P2Y receptor antagonists suramin (30 μM), pyridoxal-phosphate-6-azophenyl-2',4' disulfonic acid (PPADS; 30 μM) and reactive blue 2 (RB-2; 10 μM). Each of the antagonists significantly (p < 0.05 or 0.001) blocked uridine enhancement of NGF-stimulated neurite outgrowth (Figure 21). None of the P2Y receptor antagonists inhibited the uptake of uridine into the PCl 2 cells These results show that signaling via P2Y receptors mediates uridine induction of neurite outgrowth.
EXAMPLE 12
Phosphatidyl^ ositol (IP) signaling is stimulated by UTP and uridine
METHODS
Metabolic labeling and Pf turnover analysis [00219] Analysis of PI turnover was performed as described by (Nitsch RM et al, J
Neurochem 69: 704-12, 1997). Briefly, cells were labeled metabolically for 36 h with 1.25 microCurie (μCi)/dish of myo-[2-3H]inositol (17.0 curie/mmol; Amersham Biosciences) in serum-free MEM, washed twice with Hank's balanced salt solution (HBSS), and treated for 15 min with 10 mM lithium chloride in HBSS. Drugs were added in the presence of 10 mM lithium for 60 min at 370C. Cells were lysed with ice-coid methanol, and lipids were removed by extraction with chloroform/methanol/water (2:2:1 by volume). Labeled water-soluble inositol phosphates were separated from free [3H]inositol by ion-exchange chromatography, using AG 1-X8 columns (Bio-Rad), and IM ammonium formate and 0.1 M formic acid as eluent. Radioactivity was quantified by liquid scintillation spectrometry.
RESULTS
[00220] P2Y2, P2Y4 and P2Y6 receptors activate the phospholipase
C/diacylglycerol/inositol triphosphate (PLC/DAG/IP3) signaling pathway. To determine whether concentrations of uridine oi UTP that promote neurite outgrowth activate these receptors, NGF-differentiated PC 12 cells were labeled with [3H]-inositoI (50 μM) or UTP (10,
100 μM) for 1 hour, and IP signaling was assessed by measuring turnover of radio-labeled IP (Figure 22). Formation of IP was significantly incieased by addition of 100 μM UTP (p < 0.05) and by 50 μM uridine (p < 0.01 ). The P2Y receptor antagonist PPADS (100 μM) significantly (p < 0.05) blocked the stimulation of IP signaling by UTP. These findings indicate that UTP promotes neurite outgrowth via P2Y receptors-mediated stimulation of the IP signaling pathway.
[00221] The findings of Examples 10-12 provide a mechanism by which uridine and its metabolites stimulate neurite outgrowth: namely, by activation of P2Y receptors At least part of the action of the P2Y receptors is mediated by IP signaling Taken together, the findings from Examples 7-12 provide furthei evidence that uridine treatment can improve cognitive function by enhancing neurotransmission by multiple mechanisms: (1) enhancing neurotransmitter release; (2) acting, through CTP, as a precursor for membrane phosphatides; (3) activating, through UTP, the P2Y receptor-coupled intracellular signaling pathway Mechanisms (2) and (3) may act together to increase neurite formation.
EXAMPLE 13
UMP-supplemented diets enhance learning and memory in multiple species
MATERIALS AND EXPERIMENTAL METHODS
Morris Water Maze
[00222] Aging rats (18 months, 500 g) were fed a control diet or a diet containing 2.5%
UMP diets for six weeks. They were then shown a hidden platform in a six-foot diameter pool of water, placed somewhere in each of the four quadrants of the pool in turn, and were allowed 90 seconds in each trial to attempt to relocate the platform by swimming, and the swimming time "mean escape latency" recorded. The set of four trials was repeated on each of four consecutive days. The platform was in the same place each day. This test, known as the Morris water maze, is an indicator of spatial memory.
Food pellet learning assay
[00223] Male young adult gerbils fed control or UMP-containing chow (0, 0 1, 0.5 or
2.5%) ad lib for three weeks were tested in a radial arm maze, consisting of a central chamber with four branches primed with a small food pellet at the end of each. Before testing, animals were fasted overnight; each animal was then placed in the central chamber and allowed up to 180 seconds to find all of the pellets A shorter time required to find the pellets is indicative of improved learning and spatial memory
Working memory and reference memory assay [00224] Groups often gerbils fed control or 0.1 % UMP diet for four weeks and trained to successfully find all of the food pellets as described above were then given a modified test, in which only two arms of the maze (but always the same two) contained food pellet rewards. In this test, a working memory error is one in which a gerbil revisits an arm from which it has already taken the pellet that day. A reference memory error is one in which the gerbil enters an arm which never had food pellets (during the modified tests).
RESULTS
[00225] Previous Examples showed that orally administered uridine improves augments the ability of neural cells to function in several ways. The present Example directly shows that uridine augments cognitive function. Aging rats (18 months, 500 g) were fed a control diet or a diet containing 2.5% UMP 2Na+ for six weeks, and theii memory was tested using the Morris water maze, an indicator of spatial memory Rats administered the UMP-2Na+-fortified diet showed a statistically significant reduction in the time required to reach the location of the platform (Figure 23), indicating that UMP enhances spatial memory.
[00226] The effect of orally administered uridine upon learning and spatial memory was also examined in gerbils. Male young adult gerbils fed control or UMP-coπtaining chow (0, 0.-1, 0.5 or 2.5%) ad lib for three weeks were tested in a radial arm maze, consisting of a central chamber with four branches primed with a small food pellet at the end of each. Before testing, animals were fasted overnight; each animal was then placed in the central chamber and allowed up to 180 seconds to find all of the pellets. The reduction in time needed to find the pellets requires spatial learning UMP-supplemented diets reduced the time required for gerbils to find the pellet in a dose-dependent manner (Figure 24).
[00227] In addition, the effect of orally administered uridine on working memory and reference memory was examined. Gerbils fed a control or a 0.1% UMP diet for four weeks and trained to successfully find all of the food pellets as described above were then given a modified test, that measures working memory and reference memory Gerbils fed the UMP- supplemented diet exhibited reduced numbers of both working memory errors (Figure 25A) and reference memory errors (B). [00228] These findings directly show that (a) undine dietary supplementation improves learning and various types (spatial, working, and reference) of memory; (b) the effect is not limited to a particular species; and (c) the effect is manifested in biologically relevant models of age-impaired cognitive function.
[00229] In summary, the findings presented herein demonstrate that orally administered uridine positively affects neurological signaling, neural cell anatomy and cognitive function The findings also implicate several mechanisms by which uridine exerts its effects.
EXAMPLE 14 URIDINE AND CHOLINE INCREASE NEUROTRANSMITTER RELEASE
MATERIALS AND EXPERIMENTAL METHODS
Brain slice preparation
[00230] Male Sprague-Dawley rats, 9-1 1 months old, were anesthetized with ketamine
(85 mg/kg of body weight, intramuscularly) and were decapitated in a cold room at 4° C. Brains were rapidly removed and placed into chilled (4° C) oxygenated Kiebs buffer (1 19 5 mM NaCl, 3 3 mM KCl, 1 3 mM CaCl2, 1.2 mM MgSO4, 25 mM NaHCO3, 1 2 mM, IG-I2PO4, 1 1 mM glucose, and 0.03 mM EDTA, pH 7 4) containing 1 mM ketamine and 15 μg/ml eserine. After removal of remaining meninges and chorioid plexus. 30 μm slices of striatum, hippocampus, and coilex were immediately prepared with a Mclllwain tissue chopper, washed 3 times, and placed into custom-made superfiision chambers (Warner Instrument, Hamden, CT).
Superfiision and electrical stimulation.
[00231] Slices were equilibrated for 60 min at 37° C by superfusing the chambers with oxygenated Krebs/ketamine/eserine buffer described above at a flow rate of 0.8 ml/min Superfiision chambers contained two opposing silver mash electodes that were connected to an electrical stimulator (model S88; Grass Instruments) A custom-made polarity reversal device was used to prevent chamber polarization and also to monitor both the current and voltage 50
-microseconds after, the onset, of _each pulse to .ensure, uniform chamber resistance.. After the equilibration period, slices were depolarized by perfusion with a high-K+ (52 mM) version of the Krebs/ketamine/eserine buffer in the presence or absence of 20 μM choline, 25 μM cytidine, and/or 25 μM uridine. Perfusates were collected during the entire 2-hour period and assayed for acetylcholine Values were normalized for protein content of slices RESULTS
[00232] To determine the effect of uridine and choline on acetylcholine release, slices of striatum, hippocampus, and cortex (n=8) were incubated in the presence oi absence of choline and then depolarized, and acetylcholine release was measured. In some groups, cytidine oi uridine was added as well. Choline increased acetylcholine release whether or not uridine was also present (Figure 26).
[00233] These findings show that when neurons are repeatedly stimulated to release acetylcholine, choline increases the amount of neurotransmitter that is released, by replenishing stores of choline in membrane phospholipids (e.g. PC) The above Examples have shown that uridine augments synthesis of CDP-choline, which is then used to synthesize new PC. Together with the findings of this Example, these results show that the ability of neurons to synthesize new phospholipids, and thus repeatedly release neurotransmitters, will be increased in an additive or synergistic fashion by addition of uridine together with choline.
EXAMPLE 15 URIDINE AND CHOLINE INCREASE NEUROTRANSMITTER RELEASE
ADDITIVELY FOLLOWING REPEATED DEPOLARIZATION
[00234] Brain slices are repeatedly stimulated as described in the previous Example, in this case for 8 cycles or alternating 20-minute periods of stimulation and rest In all groups, the amount of neurotransmitter release decreases with each successive stimulation period; however, this decrease is significantly less in the presence of either uridine or choline. This effect is enhanced by the presence of both uridine and choline. Thus, uridine and choline the total amount of neurotransmitter release after repeated stimulation is increased by the presence of uridine or choline, and is further increased by the presence of uridine and choline.

Claims

What is claimed is:
1. A method of improving a cognitive function in a subject, comprising administering to said subject a uridine or a source thereof, thereby improving a cognitive function in a subject
2. A method of improving a cognitive function in a subject, comprising administering to said subject a composition, said composition comprising a uridine or a source thereof and a choline, thereby improving a cognitive function in a subject.
3 A method of treating or ameliorating a decline in a cognitive function in a subject, comprising administering a uridine or a source thereof to said subject, thereby inhibiting or preventing a decline in a cognitive function in a subject
4. A method of treating or ameliorating a decline in a cognitive function in a subject, comprising administering to said subject a composition, said composition comprising a uridine or a source thereof and a choline, thereby inhibiting or preventing a decline in a cognitive function in a subject.
5. The method of claim 3 or 4, wherein said decline is a result of a caidiovascular disease, neurodegenerative disease, oi psychiatric disease.
6. The method of claim 1 , 2, 3, or 4, wherein said cognitive function is memory, learning, intelligence, or mental fitness.
7. A method of improving-a neurological function in a subject, comprising administering to said subject a uridine or a source thereof, thereby improving a neurological function in a subject
8 A method of improving a neurological function in a subject, comprising administering to said subject a composition, said composition comprising a uridine or a source thereof and a choline, thereby improving a neurological function in a subject
9. The method of claim 7 or 8, wherein said neurological function is a synaptic transmission or a function of a neurotransmittei
10. A method of increasing a level of cytidine, cytidine triphosphate, or CDP-choline in a brain of a subject, comprising administering to said subject a uridine or a source thereof, thereby increasing a level of cytidine, cytidine triphosphate, or CDP-choline in a brain of a subject
11 A method of increasing a level of cytidine, cytidine triphosphate, or CDP-choline in a brain of a subject, comprising administering to said subject a composition, said composition comprising a uridine or a source thereof and a choline, thereby increasing a level of cytidine, cytidine triphosphate, or CDP-choline in a brain of a subject
12. A method of increasing or enhancing an ability of a brain cell or neural cell of a subject to synthesize a neurotransmitter, comprising administering to said subject a uridine or a source thereof, thereby increasing or enhancing an ability of a brain cell or neural cell of a subject to synthesize a neurotransmitter.
13. A method of increasing or enhancing an ability of a brain cell or neural cell of a subject to synthesize a neurotransmitter, comprising administering to said subject a composition, said composition comprising a uridine or a source thereof and a choline,- thereby increasing or enhancing an ability of a brain cell or neural cell of a subject to synthesize a neurotransmitter
14, A method of increasing a level of a neurotransmitter in a synapse, comprising contacting a neural cell adjacent to said synapse with a uridine or a source thereof, whereby said uridine enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a level of a neurotransmitter in a synapse.
15. A method of inci easing a level of a neurotransmitter in a synapse, comprising contacting a neural cell adjacent to said synapse with a composition, said composition comprising a uridine or a source thereof and a choline, whereby said composition enhances synthesis of a phospholipid or a precursor thereof, thereby increasing a level of a neurotransmitter in a synapse, comprising
16 The method of claim 9, 1 2, 13, 14, or 15, wherein said neurotransmitter is acetylcholine.
17. A method of stimulating oi enhancing a synthesis of a membrane of a biain cell or neural cell of a subject, comprising administering to said subject a uridine or a source theieof, thereby stimulating or enhancing a synthesis of a membrane of a brain cell or a neural cell of a subject.
18 A method of stimulating or enhancing a synthesis of a membrane of a brain cell or neural cell of a subject, comprising administering to said subject a composition, said composition comprising a uiidine or a source thereof and a choline, thereby stimulating or enhancing a synthesis of a membrane of a brain cell or a neural cell of a subject.
19. A method of stimulating or enhancing an outgrowth of a neurite of a neural cell of a - - subject,- comprising administering-to said subject a- uridine or a source thereof, thereby stimulating or enhancing an outgrowth of a neurite of a neural cell of a subject.
20. A method of stimulating or enhancing an outgrowth of a neurite of a neural cell of a subject, comprising administering to said subject a composition, said composition comprising a undine or a source thereof and a choline, thereby stimulating or enhancing an outgrowth of a neurite of a neural cell of a subject,
21- The method of claim 17, 18, 19, or 20, whereby said composition enhances a -production of a phospholipid, thereby stimulating oi enhancing said synthesis of a membrane or outgrowth of a neurite
22. The method of any of claims 1-21, wherein said uridine is uridine-5 '-monophosphate, uridine-5'-diphosphate, or uridine-5'-triphosphate.
23 The method of any of claims 1-21 , wherein said source is citicoline (CDP-choline)
24. The method of any of claims 2, 4, 5, 6, 8, 9, 1 1 , 13, 15, 16, 18, 20, 21, 22, or 23, wherein said choline is a choline salt
25. The method of any of claims 1-1 1 , 22, or 23, whereby said uridine or a metabolite thereof mediates its effect by stimulating a P2Y receptor in a neural cell or brain cell of said subject
26 The method of any of claims 12-21 , whereby said uridine or a metabolite thereof mediates its effect by stimulating a P2Y receptor in said neural cell or brain cell.
27. The method of either of claims 25 or 26, wherein said metabolite is uridine-5"- triphosphatc.
EP05796529A 2004-09-15 2005-09-13 Compositions containing uridine, and methods utilizing same Ceased EP1802314A4 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US10/941,025 US20050203053A1 (en) 1999-07-30 2004-09-15 Uridine administration improves phosphatide synthesis, synaptic transmission and cogntive function
US10/944,269 US8143234B2 (en) 1998-07-31 2004-09-20 Uridine administration improves phosphatide synthesis, synaptic transmission and cognitive function
US10/972,777 US8314064B2 (en) 1998-07-31 2004-10-26 Uridine administration stimulates membrane production
PCT/US2005/032312 WO2006031683A2 (en) 2004-09-15 2005-09-13 Compositions containing uridine, and methods utilizing same

Publications (2)

Publication Number Publication Date
EP1802314A2 true EP1802314A2 (en) 2007-07-04
EP1802314A4 EP1802314A4 (en) 2011-02-23

Family

ID=36060581

Family Applications (1)

Application Number Title Priority Date Filing Date
EP05796529A Ceased EP1802314A4 (en) 2004-09-15 2005-09-13 Compositions containing uridine, and methods utilizing same

Country Status (6)

Country Link
EP (1) EP1802314A4 (en)
JP (2) JP2008513453A (en)
CN (1) CN102600199A (en)
AU (1) AU2005285090A1 (en)
CA (1) CA2579851C (en)
WO (1) WO2006031683A2 (en)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8143234B2 (en) 1998-07-31 2012-03-27 Massachusetts Institute Of Technology Uridine administration improves phosphatide synthesis, synaptic transmission and cognitive function
US8518882B2 (en) 1998-07-31 2013-08-27 Massachusetts Institute Of Technology Methods and compositions for ameliorating or inhibiting decline in memory or intelligence or improving same
US8314064B2 (en) 1998-07-31 2012-11-20 Massachusetts Institute Of Technology Uridine administration stimulates membrane production
CN102600199A (en) * 2004-09-15 2012-07-25 麻省理工学院 Methods for increasing cytidine levels in vivo and treating cytidine-dependent human diseases
WO2009002148A1 (en) 2007-06-27 2008-12-31 N.V. Nutricia Food composition for prodromal dementia patients
PT2170316E (en) * 2007-06-26 2014-01-30 Nutricia Nv Improving memory in subjects with mini-mental state examination of 24-26
WO2009002145A1 (en) 2007-06-26 2008-12-31 N.V. Nutricia Lipid composition for improving function of brain functioning
WO2009002146A1 (en) 2007-06-26 2008-12-31 N.V. Nutricia Supporting activities of daily living
WO2009057994A1 (en) 2007-11-02 2009-05-07 N.V. Nutricia Unit dosage for brain health
MX2010006829A (en) 2007-12-20 2010-09-30 Nutricia Nv Liquid nucleotides/nucleosides-containing product.
WO2012125020A1 (en) * 2011-03-14 2012-09-20 N.V. Nutricia Method for treating neurotrauma
WO2014160502A1 (en) * 2013-03-13 2014-10-02 Tufts University Uridine nucleoside derivatives, compositions and methods of use
CN106822393A (en) * 2017-02-17 2017-06-13 福建康是美生物科技有限公司 A kind of health-caring capsule for strengthening memory
AU2019215105A1 (en) * 2018-02-01 2020-08-13 Pharma Cinq, Llc Compositions and devices for systemic delivery of uridine
CN115120607A (en) * 2022-06-07 2022-09-30 珍奥集团股份有限公司 Application of nucleotide mixture in preparation for preventing and treating Alzheimer's disease
WO2024058276A1 (en) * 2022-09-16 2024-03-21 ヤマサ醤油株式会社 Agent and method for improving composite memory of normal person
CN115737666B (en) * 2022-11-24 2024-03-26 暨南大学 Application of uridine diphosphate glucose in preparation of anti-aging products

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997045127A1 (en) * 1996-05-28 1997-12-04 Polifarma S.P.A. Uridine-comprising therapeutic active agent for treatment of neurodegenerative disorders
WO2000006174A1 (en) * 1998-07-31 2000-02-10 Massachusetts Institute Of Technology Methods for increasing cytidine levels in vivo and treating cytidine-dependent human diseases
US20030114415A1 (en) * 2001-12-14 2003-06-19 Wurtman Richard J. Compositions and methods for treating and preventing memory impairment using citicoline
WO2005079250A2 (en) * 2004-02-13 2005-09-01 Cornell Research Foundation, Inc. Purines are self-renewal signals for neural stem cells, and purine receptor antagonists promote neuronal and glial differentiation therefrom
WO2005112635A2 (en) * 2004-05-13 2005-12-01 Massachusetts Institute Of Technology Uridine effects on dopamine release

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3011A (en) * 1843-03-21 Improvement in water-wheels
BR9709379A (en) * 1996-05-28 2000-01-11 Polifarma Spa Use of uridine, and, composition for the treatment of disorders of the nervous system due to the selective degeneration of neuronal or glial cells in mammals.
US6472378B2 (en) * 1998-08-31 2002-10-29 Pro-Neuron, Inc. Compositions and methods for treatment of mitochondrial diseases
JP2001233776A (en) * 2000-02-25 2001-08-28 Yamasa Shoyu Co Ltd Agent for improving lowering of learning and memorizing ability and its use
CN102600199A (en) * 2004-09-15 2012-07-25 麻省理工学院 Methods for increasing cytidine levels in vivo and treating cytidine-dependent human diseases

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997045127A1 (en) * 1996-05-28 1997-12-04 Polifarma S.P.A. Uridine-comprising therapeutic active agent for treatment of neurodegenerative disorders
WO2000006174A1 (en) * 1998-07-31 2000-02-10 Massachusetts Institute Of Technology Methods for increasing cytidine levels in vivo and treating cytidine-dependent human diseases
US20030114415A1 (en) * 2001-12-14 2003-06-19 Wurtman Richard J. Compositions and methods for treating and preventing memory impairment using citicoline
WO2005079250A2 (en) * 2004-02-13 2005-09-01 Cornell Research Foundation, Inc. Purines are self-renewal signals for neural stem cells, and purine receptor antagonists promote neuronal and glial differentiation therefrom
WO2005112635A2 (en) * 2004-05-13 2005-12-01 Massachusetts Institute Of Technology Uridine effects on dopamine release

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CACABELOS R ET AL: "THERAPEUTIC EFFECTS OF CDP-CHOLINE IN ALZHEIMER'S DISEASE COGNITION, BRAIN MAPPING, CEREBROVASCULAR HEMODYNAMICS, AND IMMUNE FACTORS", ANNALS OF THE NEW YORK ACADEMY OF SCIENCES, NEW YORK ACADEMY OF SCIENCES, NEW YORK, NY, US, 1 January 1996 (1996-01-01), pages 399-403, XP008065562, ISSN: 0077-8923, DOI: DOI:10.1111/J.1749-6632.1996.TB34452.X *
DE BRUIN N M W J ET AL: "Combined uridine and choline administration improves cognitive deficits in spontaneously hypertensive rats", NEUROBIOLOGY OF LEARNING AND MEMORY, SAN DIEGO, US, vol. 80, no. 1, 1 July 2003 (2003-07-01), pages 63-79, XP002603409, ISSN: 1074-7427, DOI: DOI:10.1016/S1074-7427(03)00024-8 [retrieved on 2003-04-11] *
See also references of WO2006031683A2 *
SPIERS P A ET AL: "CITICOLINE IMPROVES VERBAL MEMORY IN AGING", ARCHIVES OF NEUROLOGY, AMERICAN MEDICAL ASSOCIATION, CHICAGO, IL, US, vol. 53, no. 5, 1 May 1996 (1996-05-01), pages 441-448, XP008028412, ISSN: 0003-9942 *

Also Published As

Publication number Publication date
AU2005285090A1 (en) 2006-03-23
EP1802314A4 (en) 2011-02-23
CN102600199A (en) 2012-07-25
JP2013064026A (en) 2013-04-11
JP2008513453A (en) 2008-05-01
WO2006031683A3 (en) 2006-12-21
CA2579851C (en) 2018-09-04
JP6141643B2 (en) 2017-06-07
CA2579851A1 (en) 2006-03-23
WO2006031683A2 (en) 2006-03-23

Similar Documents

Publication Publication Date Title
CA2579851C (en) Use of uridine for improving cognitive and neurological functions
US8518882B2 (en) Methods and compositions for ameliorating or inhibiting decline in memory or intelligence or improving same
US20070004670A1 (en) Compositions containing citicoline, and methods of use thereof
US20050203053A1 (en) Uridine administration improves phosphatide synthesis, synaptic transmission and cogntive function
US8030294B2 (en) Compounds for the treatment of psychiatric or substance abuse disorders
US20060069061A1 (en) Compositions containing uridine and choline, and methods utilizing same
Wang et al. Dietary uridine-5′-monophosphate supplementation increases potassium-evoked dopamine release and promotes neurite outgrowth in aged rats
US8143234B2 (en) Uridine administration improves phosphatide synthesis, synaptic transmission and cognitive function
US8691775B2 (en) Use of drugs that activate P2Y receptors to enhance synaptogenesis
JP2003517437A (en) Methods for increasing cytidine levels in vivo and treating cytidine-dependent human diseases
JP2008502705A (en) Compounds and methods for treating seizures and seizure disorders
US8314064B2 (en) Uridine administration stimulates membrane production
AU2011253660B2 (en) Compositions containing uridine, and methods utilizing same
Villanueva-Castillo et al. The role of purinergic P2Y12 and P2Y13 receptors in ADPβS-induced inhibition of the cardioaccelerator sympathetic drive in pithed rats

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20070416

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MASSACHUSETTS INSTITUTE OF TECHNOLOGY

DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20110120

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MASSACHUSETTS INSTITUTE OF TECHNOLOGY

17Q First examination report despatched

Effective date: 20130510

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

REG Reference to a national code

Ref country code: DE

Ref legal event code: R003

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20210716