COMPOSITIONS AND METHODS CONTAINING REDUCED NICOTINAMIDE RIBOSIDE FOR PREVENTION AND TREATMENT OF NEUROLOGICAL DISEASES AND CONDITIONS

Information

  • Patent Application
  • 20230114280
  • Publication Number
    20230114280
  • Date Filed
    March 09, 2021
    3 years ago
  • Date Published
    April 13, 2023
    a year ago
Abstract
The present invention provides compounds and compositions containing reduced nicotinamide riboside for use in methods of prevention and/or treatment of neurological disease and/or conditions. In one embodiment of the invention, said compounds and compositions of the invention maintain or improve brain function, in particular brain energy deficits. In another embodiment of the invention, compounds and compositions of the invention improve neurological recovery and regeneration after injury or surgery. In another embodiment of the invention, compounds and compositions of the invention may be used in methods to prevent and/or treat neurological diseases and/or conditions and/or recovery after injury or surgery.
Description
FIELD OF THE INVENTION

The present invention provides compounds and compositions containing reduced nicotinamide riboside for use in methods of prevention and/or treatment of neurological disease and/or conditions. In one embodiment of the invention, said compounds and compositions of the invention maintain or improve brain function, in particular brain energy deficits. In another embodiment of the invention, compounds and compositions of the invention improve neurological recovery and regeneration after injury or surgery. In another embodiment of the invention, compounds and compositions of the invention may be used in methods to prevent and/or treat neurological diseases and/or conditions and/or recovery after injury or surgery.


BACKGROUND TO THE INVENTION

Brain tissue consumes a large amount of energy in proportion to its volume. In an average healthy subject, the brain obtains most of its energy from oxygen-dependent metabolism of glucose. Typically, the majority of the brain’s energy is used to help neurons or nerve cells send signals and the remaining energy is used for cell-health maintenance. A deficiency in brain energy, for example caused by impairment of glucose utilisation, can result in neuronal hyperactivity, seizures and cognitive impairments.


Examples of brain energy deficiency conditions or diseases include: migraine, memory disorder, age-related memory disorder, brain injury, neurorehabilitation, stroke and post-stroke, amyloid lateral sclerosis, multiple sclerosis, cognitive impairment, mild cognitive impairment (MCI), cognitive impairment post-intensive care, age-induced cognition impairment, Alzheimer’s disease, Parkinson’s disease, Huntingdon’s disease, inherited metabolic disorders (such as glucose transporter type 1 deficiency syndrome and pyruvate dehydrogenase complex deficiency), bipolar disorder, schizophrenia, epilepsy, stress and/or motivational performance.


Brain energy deficiency conditions may also occur in the absence of disease during conditions of high performance or psychological stress.


NAD+ plays an important role in neurological development, regeneration, aging and disease. NAD+ mediates multiple biological processes in brains, such as neurotransmission and learning and memory. NAD+ may also mediate brain aging and the tissue damage in various brain illnesses. NADH can be transported across the plasma membranes of astrocytes, and NAD+ administration can markedly decrease ischemic brain injury (Ying et al. 2007). Lower NAD+ levels may be deleterious for brain health while higher NAD+ levels are known to augment brain health. Therefore, there is an urgent unmet need to address neurological disease and/or conditions with new compounds, compositions and methods of prevention and/or treatment which influence NAD+.


SUMMARY OF THE INVENTION

The present invention provides compounds and compositions for use in methods of prevention and/or treatment of neurological conditions and diseases.


In an embodiment, the composition is selected from the group consisting of: a food or beverage product, a food supplement, an oral nutritional supplement (ONS), a medical food, and combinations thereof.


In another embodiment, the present invention provides a method for increasing intracellular nicotinamide adenine dinucleotide (NAD+) in a subject, the method comprising administering a compound or composition of the invention consisting of administering a reduced nicotinamide riboside to the subject in an amount effective to increase NAD+ biosynthesis.


In a further embodiment, as a precursor of NAD+ biosynthesis, reduced nicotinamide riboside, can increase in NAD+ biosynthesis and provide one or more benefits to neurological function.


In another embodiment, as a precursor of NAD+ biosynthesis, reduced nicotinamide riboside, can increase in NAD+ biosynthesis and provide one or more benefits to brain function.


In another embodiment, the present invention provides a unit dosage form of a composition consisting of reduced nicotinamide riboside, the unit dosage form contains an effective amount of the reduced nicotinamide riboside to increase NAD+ biosynthesis.


In one embodiment of the invention, the composition containing reduced nicotinamide riboside is provided to maintain or increase brain function in a subject.


In yet another embodiment of the invention, the composition containing reduced nicotinamide riboside is provided to enhance neurological recovery after injury or surgery.


In another embodiment of the invention, the composition is a nutritional composition selected from a: food or beverage product, including food additives, food ingredients, functional foods, dietary supplements, medical foods, nutraceuticals, oral nutritional supplements (ONS) or food supplements.


In another embodiment of the invention, the composition is a nutritional composition containing reduced nicotinamide riboside wherein increased neurological function in the brain is measured by, for example, suitable neurological and cognitive tests, brain image analysis and clinical examination.


DETAILED DESCRIPTION OF THE INVENTION
Definitions

All percentages expressed herein are by weight of the total weight of the composition unless expressed otherwise. As used herein, “about,” “approximately” and “substantially” are understood to refer to numbers in a range of numerals, for example the range of -10% to +10% of the referenced number, preferably -5% to +5% of the referenced number, more preferably —1% to +1 % of the referenced number, most preferably -0.1 % to +0.1 % of the referenced number.


All numerical ranges herein should be understood to include all integers, whole or fractions, within the range. Moreover, these numerical ranges should be construed as providing support for a claim directed to any number or subset of numbers in that range. For example, a disclosure of from 1 to 10 should be construed as supporting a range of from 1 to 8, from 3 to 7, from 1 to 9, from 3.6 to 4.6, from 3.5 to 9.9, and so forth.


As used in this invention and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a component” or “the component” includes two or more components.


The words “comprise,” “comprises” and “comprising” are to be interpreted inclusively rather than exclusively. Likewise, the terms “include,” “including” and “or” should all be construed to be inclusive, unless such a construction is clearly prohibited from the context. Nevertheless, the compositions disclosed herein may lack any element that is not specifically disclosed herein. Thus, a disclosure of an embodiment using the term “comprising” includes a disclosure of embodiments “consisting essentially of” and “consisting of” the components identified. Any embodiment disclosed herein can be combined with any other embodiment disclosed herein.


Where used herein, the terms “example” and “such as,” particularly when followed by a listing of terms, are merely exemplary and illustrative and should not be deemed to be exclusive or comprehensive. As used herein, a condition “associated with” or “linked with” another condition means the conditions occur concurrently, preferably means that the conditions are caused by the same underlying condition, and most preferably means that one of the identified conditions is caused by the other identified condition.


The terms “food,” “food product” and “food composition” mean a product or composition that is intended for ingestion by an individual such as a human and provides at least one nutrient to the individual. A food product typically includes at least one of a protein, a lipid, a carbohydrate and optionally includes one or more vitamins and minerals. The term “beverage” or “beverage product” means a liquid product or liquid composition that is intended to be ingested orally by an individual such as a human and provides at least one nutrient to the individual.


The compositions of the present disclosure, including the many embodiments described herein, can comprise, consist of, or consist essentially of the elements disclosed herein, as well as any additional or optional ingredients, components, or elements described herein or otherwise useful in a diet.


As used herein, the term “isolated” means removed from one or more other compounds or components with which the compound may otherwise be found, for example as found in nature. For example, “isolated” preferably means that the identified compound is separated from at least a portion of the cellular material with which it is typically found in nature. In an embodiment, an isolated compound is free from any other compound.


“Prevention” includes reduction of risk, incidence and/or severity of a condition or disorder. The terms “treatment,” “treat” and “to alleviate” include both prophylactic or preventive treatment (that prevent and/or slow the development of a targeted pathologic condition or disorder) and curative, therapeutic or disease-modifying treatment, including therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder; and treatment of patients at risk of contracting a disease or suspected to have contracted a disease, as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition. The term does not necessarily imply that a subject is treated until total recovery. The terms “treatment” and “treat” also refer to the maintenance and/or promotion of health in an individual not suffering from a disease but who may be susceptible to the development of an unhealthy condition. The terms “treatment,” “treat” and “to alleviate” are also intended to include the potentiation or otherwise enhancement of one or more primary prophylactic or therapeutic measure. The terms “treatment,” “treat” and “to alleviate” are further intended to include the dietary management of a disease or condition or the dietary management for prophylaxis or prevention a disease or condition. A treatment can be patient- or doctor-related.


The term “unit dosage form,” as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of the composition disclosed herein in an amount sufficient to produce the desired effect, in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for the unit dosage form depend on the particular compounds employed, the effect to be achieved, and the pharmacodynamics associated with each compound in the host.


As used herein, an “effective amount” is an amount that prevents a deficiency, treats a disease or medical condition in an individual, or, more generally, reduces symptoms, manages progression of the disease, or provides a nutritional, physiological, or medical benefit to the individual. The relative terms “improve,” “increase,” “enhance,” “promote” and the like refer to the effects of the composition disclosed herein, namely a composition comprising reduced nicotinamide riboside, relative to a composition not having nicotinamide riboside but otherwise identical. As used herein, “promoting” refers to enhancing or inducing relative to the level before administration of the composition disclosed herein.


As used herein “reduced nicotinamide riboside” may also be known as protonated nicotinamide riboside, dihydronicotinamide riboside, dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide, or 1-(beta-D-ribofuranosyl)-dihydronicotinamide. A description of the synthesis of reduced nicotinamide riboside is given in Example 1. The location of the protonation site can give rise to different forms of “reduced nicotinamide riboside”. For example: 1,4-dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide; 1,2-dihydro-1 -beta-D-ribofuranosyl-3-pyridinecarboxamide; and 1,6-dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide (Makarov and Migaud, 2019).


Neurological Diseases and Conditions

As used herein, the term “neurological condition” refers to a disorder of the nervous system. Neurological conditions may result from damage to the brain, spinal column or nerves, caused by illness or injury. Non-limiting examples of the symptoms of a neurological condition include paralysis, muscle weakness, poor coordination, loss of sensation, seizures, confusion, pain and altered levels of consciousness. An assessment of the response to touch, pressure, vibration, limb position, heat, cold, and pain as well as reflexes can be performed to determine whether the nervous system is impaired in a subject.


Some neurological conditions are life-long, and the onset can be experienced at any time. Other neurological conditions, such as cerebral palsy, are present from birth. Some neurological conditions, such as Duchenne muscular dystrophy, commonly appear in early childhood, while other neurological conditions, such as Alzheimer’s disease and Parkinson’s disease, affect mainly older people. Some neurological conditions have a sudden onset due to injury or illness, such as a head injury or stroke, or cancers of the brain and spine.


In an embodiment, the neurological condition is the result of traumatic damage to the brain. Additionally or alternatively, the neurological condition is the result of an energy deficiency in the brain or in the muscles.


Examples of neurological conditions include migraine, memory disorder, age-related memory disorder, brain injury, neurorehabilitation, stroke and post-stroke, amyloid lateral sclerosis, multiple sclerosis, cognitive impairment, mild cognitive impairment (MCI), cognitive impairment post-intensive care, age-induced cognition impairment, Alzheimer’s disease, Parkinson’s disease, Huntingdon’s disease, inherited metabolic disorders (such as glucose transporter type 1 deficiency syndrome and pyruvate dehydrogenase complex deficiency), bipolar disorder, schizophrenia, and/or epilepsy.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat neurological conditions listed above, in particular, to maintain or improve brain or nervous system function.


Migraine

A migraine is an intense headache accompanied by other symptoms such as nausea (feeling sick), visual problems and an increased sensitivity to light or sound. A migraine may be preceded by an aura; the main symptoms of an aura are visual problems such as blurred vision (difficulty focusing), blind spots, flashes of light, or a zigzag pattern moving from the central field of vision towards the edge.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat migraine or its neurological symptoms.


Stroke

Strokes (also known as cerebrovascular accident (CVA) and cerebrovascular insult (CVI)) occur when there is poor blood flow to the brain resulting in cell death. There are two main types of stroke: ischemic (due to lack of blood flow) and haemorrhagic (due to bleeding). Strokes result in part of the brain not functioning properly. The signs and symptoms of a stroke may include an inability to move or feel on one side of the body, problems understanding or speaking, feeling like the world is spinning, or loss of vision to one side. The signs and symptoms often appear soon after the stroke has occurred.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat stroke or recovery from stroke.


Amytrophic Lateral Sclerosis

Amyotrophic lateral sclerosis (ALS) (also known as Lou Gehrig’s disease, Charcot disease and motor neuron disease), involves the death of neurons responsible for controlling voluntary muscles. ALS is characterized by stiff muscles, muscle twitching, and gradually worsening weakness due to muscle wasting; this results in difficulty speaking, swallowing, and eventually breathing.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat ALS or its neurological symptoms.


Multiple Sclerosis

Multiple sclerosis (MS) affects the nerves in the brain and spinal cord, causing a wide range of symptoms including problems with muscle movement, problems with mobility and balance, numbness and tingling, blurring of vision (typically there is loss of vision in one eye) and fatigue.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat MS or its neurological symptoms.


Parkinson’s Disease

Parkinson’s disease is a degenerative disorder of the central nervous system mainly affecting the motor system. In the early course of the disease, the most obvious symptoms are movement-related; these include tremor at rest, rigidity, slowness of movement and difficulty with walking and gait. Later in the course of the disease, thinking and behavioral problems may arise, with dementia commonly occurring in the advanced stages of the disease. Other symptoms include depression, sensory, sleep and emotional problems.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat Parkinson’s disease or its neurological symptoms.


Alzheimer’s Disease

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder. Alzheimer’s disease is the most common cause of dementia. Symptoms include memory loss and difficulties with thinking, problem-solving or language. The mini mental state examination (MMSE) is an example of one of the tests used to diagnose Alzheimer’s disease.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat AD or its neurological symptoms.


Huntington’s Disease

Huntington’s disease is an inherited condition that damages certain nerve cells in the brain. Huntington’s disease affects muscle coordination and leads to mental decline and behavioral symptoms. The earliest symptoms are often subtle problems with mood or cognition. A general lack of coordination and an unsteady gait often follow. As the disease advances, uncoordinated, jerky body movements become more apparent, along with a decline in mental abilities and behavioral symptoms. Physical abilities gradually worsen until coordinated movement becomes difficult. Mental abilities generally decline into dementia.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat Huntington’s disease or its neurological symptoms.


Inherited Metabolic Disorders Affecting the Brain and Nervous System

Inherited metabolic disorders are a range of diseases caused by defective genes. Typically the defective gene(s) results in a defect in an enzyme or in a transport protein which results in a block in the way that a compound is processed by the body such that there is a toxic accumulation of the compound. Inherited metabolic disorders can affect any organ and usually affect more than one. Symptoms often tend to be nonspecific and usually relate to major organ dysfunction or failure. The onset and severity of a metabolic disorder may be exacerbated by environmental factors, such as diet and concurrent illness.


GLUT 1 Deficiency Syndrome

Glucose transporter type 1 (Glutl) deficiency syndrome is a genetic metabolic disorder involving the GLUT1 protein which transports glucose across the blood-brain barrier or the boundary separating tiny blood vessels from brain tissue. The most common symptom is seizures (epilepsy), which usually begin within the first few months of life. Additional symptoms that can occur include varying degrees of cognitive impairment and movement disorders characterized by ataxia, dystonia, and chorea. Glutl deficiency syndrome may be caused by mutations in the SLC2A1 gene which produce GLUT1 protein.


Pyruvate Dehydrogenase Complex Deficiency

Pyruvate dehydrogenase complex deficiency (pyruvate dehydrogenase deficiency or PDCD) is a neurodegenerative disorder associated with abnormal mitochondrial metabolism and disrupted carbohydrate metabolism. PDCD is characterized by the buildup of lactic acid in the body and a variety of neurological problems. Signs and symptoms of this condition usually first appear shortly after birth, and they can vary widely among affected individuals. The most common feature is a potentially life-threatening buildup of lactic acid (lactic acidosis), which can cause nausea, vomiting, severe breathing problems, and an abnormal heartbeat. Other symptoms include: neurological problems; delayed development of mental abilities and motor skills such as sitting and walking; intellectual disability; seizures; weak muscle tone (hypotonia); poor coordination, and difficulty walking. Some affected individuals have abnormal brain structures, such as underdevelopment of the tissue connecting the left and right halves of the brain (corpus callosum), wasting away (atrophy) of the exterior part of the brain known as the cerebral cortex, or patches of damaged tissue (lesions) on some parts of the brain.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat inherited metabolic diseases or conditions affecting the brain and/or nervous system.


Psychogenic Conditions and Disorders

Psychogenic conditions and diseases relate to emotional or mental stressors which may affect brain function. Psychogenic disorders are divided into: (i) dissociation (with memory, consciousness and self-identity impairment), and (ii) disturbances with somatizations, divided into somatoform (unconscious), factitious (voluntary search for patient’s role) and malingering (searching for material gain). Normal activity in certain brain areas of the motor or sensory cortex is blocked by other brain areas related to emotional integration in the anterior cingular and orbitofrontal cortex.


Bipolar Disorder

Bipolar disorder is a brain disorder that causes unusual shifts in mood, energy, activity levels, and the ability to carry out day-to-day tasks. Bipolar disorder is characterized by periods of elevated mood and periods of depression. Bipolar disorder can be diagnosed using the guidelines from the Diagnostic and Statistical Manual of Mental Disorders (DSM) or the World Health Organization’s International Statistical Classification of Diseases and Related Health Problems.


Schizophrenia

Schizophrenia is a chronic, severe, and disabling brain disorder in which individuals interpret reality abnormally. Schizophrenia may result in some combination of hallucinations, hearing voices, delusions, and extremely disordered thinking and behavior. Schizophrenia can be diagnosed using the guidelines from the Diagnostic and Statistical Manual of Mental Disorders (DSM) or the World Health Organization’s International Statistical Classification of Diseases and Related Health Problems.


Epilepsy

Epilepsy is a neurological disorder in which nerve cell activity in the brain becomes disrupted, causing seizures or periods of unusual behavior, sensations and sometimes loss of consciousness.


Stress

In biology and psychology, the term “stress” refers to the consequence of the failure of a human or other animal to respond appropriately to physiological, emotional, or physical threats, whether actual or imagined. The psychobiological features of stress may present as manifestations of oxidative stress, i.e., an imbalance between the production and manifestation of reactive oxygen species and the ability of a biological system readily to detoxify the reactive intermediates or to repair the resulting damage. Disturbances in the normal redox state of tissues can cause toxic effects through the production of peroxides and free radicals that damage all of the components of the cell, including proteins, lipids, and DNA. Some reactive oxidative species can even act as messengers through a phenomenon called “redox signaling.”


Motivational Performance or Mental Energy

“Motivational performance” is synonymous with the terms “mental energy” and related terms of “volition”, “will-power”, “time-on-task”, “persistence”, “self-control”, “sustained effort”, and “self-efficacy”. All these terms relate to a person’s drive to initiate and do things. Motivational performance is linked to subjectively perceived self-efficacy and well-being.


Motivational performance describes the subjective perception of mental resources available, which in turn is linked to cognitive functioning. For example, motivational performance is reduced in states of depression and anxiety. Measurement of “motivational performance” can be by both motor tasks and cognitive tasks. Typically, these motor tasks and cognitive tasks are performed under incentivized conditions, meaning that individuals get an incentive depending on their performance of the task.


It may be appreciated that the compounds, compositions and methods of the present invention may be beneficial to prevent and/or treat psychogenic disease conditions listed above and other conditions related to stress and motivational performance, in particular, to maintain or improve brain or nervous system function.


The terms “cognitive impairment” and “cognition impairment” refer to disorders that give rise to impaired cognition, in particular disorders that primarily affect learning, memory, perception, and/or problem solving.


Cognitive impairment may occur in a subject after intensive care. Cognitive impairment may occur as part of the ageing process, e.g. mild cognitive impairment (MCI).


The term “cognition” refers to the set of all mental abilities and processes, including knowledge, attention, memory and working memory, judgment and evaluation, reasoning and “computation”, problem solving and decision making, comprehension and production of language. Levels of and improvements in cognition can be readily assessed by the skilled person using any suitable neurological and cognitive tests that are known in the art, including cognitive tests designed to assess speed of information processing, executive function and memory. Suitable example tests include Mini Mental State Examination (MMSE), Cambridge Neuropsychological Test Automated Battery (CANTAB), Alzheimer’s Disease Assessment Scale-cognitive test (ADAScog), Wisconsin Card Sorting Test, Verbal and Figural Fluency Test and Trail Making Test, Wechsler Memory scale (WMS), immediate and delayed Visual Reproduction Test (Trahan et al. Neuropsychology, 1988 19(3) p. 173-89), the Rey Auditory Verbal Learning Test (RAVLT) (Ivnik, RJ. et al. Psychological Assessment: A Journal of Consulting and Clinical Psychology, 1990 (2): p. 304-312), electroencephalography (EEG), magnetoencephalography (MEG), Positron Emission Tomography (PET), Single Photon Emission Computed Tomography (SPECT), Magnetic Resonance Imaging (MRI), functional Magnetic Resonance Imaging (fMRI), computerized tomography and long-term potentiation.


EEG, a measure of electrical activity of the brain, is accomplished by placing electrodes on the scalp at various landmarks and recording greatly amplified brain signals. MEG is similar to EEG in that it measures the magnetic fields that are linked to electrical fields. MEG is used to measure spontaneous brain activity, including synchronous waves in the nervous system.


PET provides a measure of oxygen utilisation and/or glucose metabolism. In this technique, a radioactive positron-emitting tracer is administered, and tracer uptake by the brain is correlated with brain activity. These tracers emit gamma rays which are detected by sensors surrounding the head, resulting in a 3D map of brain activation. As soon as the tracer is taken up by the brain, the detected radioactivity occurs as a function of regional cerebral blood flow. During activation, an increase in cerebral blood flow and neuronal glucose metabolism can be detected within seconds.


Suitable analysis can also be based on neuropsychiatric testing, clinical examinations and individual complaints of loss of cognitive function (e.g. subjective memory loss). Further suitable tests may be based on assessments of locomotion, memory and attention, seizure susceptibility, and social interaction and/or recognition.


Memory disorders are the result of neurological damage to the brain structures such that the storage, retention and recollection of memories are hindered. Memory disorders can be progressive with age (e.g. Alzheimer’s disease), or they can be immediately resulting, for example, from a head injury. Levels of and improvements in memory disorders can be readily assessed by the skilled person using any suitable tests that are known in the art such as Alzheimer’s Disease Assessment Scale-cognitive test (ADAScog), Mini Mental State Examination (MMSE), computerized tomography (CT) scan, Magnetic Resonance Imaging (MRI), Single Photon Emission Computed Tomography (SPECT), Positron Emission Tomography (PET), and electroencephalography (EEG).


EMBODIMENTS

The present invention provides compounds and compositions containing reduced nicotinamide riboside. Another aspect of the present invention is a unit dosage form of a composition consisting of reduced nicotinamide riboside, and the unit dosage form contains the reduced nicotinamide riboside in an amount effective to increase intracellular NAD+ in subject in need thereof.


The increase in NAD+ biosynthesis can provide one or more benefits to the individual, for example a human (e.g., a human undergoing medical treatment), a pet or a horse (e.g., a pet or horse undergoing medical treatment), or cattle or poultry (e.g., cattle or poultry being used in agriculture) with respect to prevention or treatment of neurological disease and/or condition..


For non-human mammals such as rodents, some embodiments comprise administering an amount of the composition that provides 1.0 mg to 1.0 g of the reduced nicotinamide riboside / kg of body weight of the non-human mammal, preferably 10 mg to 500 mg of the reduced nicotinamide riboside / kg of body weight of the non-human mammal, more preferably 25 mg to 400 mg of the reduced nicotinamide riboside / kg of body weight of the mammal, most preferably 50 mg to 300 mg of the reduced nicotinamide riboside / kg of body weight of the non-human mammal.


For humans, some embodiments comprise administering an amount of the composition that provides 1.0 mg to 10.0 g of the reduced nicotinamide riboside / kg of body weight of the human, preferably 10 mg to 5.0 g of the reduced nicotinamide riboside / kg of body weight of the human, more preferably 50 mg to 2.0 g of the reduced nicotinamide riboside / kg of body weight of the human, most preferably 100 mg to 1.0 g of the reduced nicotinamide riboside / kg of body weight of the human.


In some embodiments, at least a portion of the reduced nicotinamide riboside is isolated from natural plant sources. Additionally or alternatively, at least a portion of reduced nicotinamide riboside can be chemically synthesized. For example, according to Example 1 described below.


As used herein, a “composition consisting essentially of reduced nicotinamide riboside” contains reduced nicotinamide riboside and does not include, or is substantially free of, or completely free of, any additional compound that affects NAD+ production other than the “reduced nicotinamide riboside”. In a particular non-limiting embodiment, the composition consists of the reduced nicotinamide riboside and an excipient or one or more excipients.


In some embodiments, the composition consisting essentially of reduced nicotinamide riboside is optionally substantially free or completely free of other NAD+ precursors, such as nicotinamide riboside.


As used herein, “substantially free” means that any of the other compounds present in the composition is no greater than 1.0 wt.% relative to the amount of reduced nicotinamide riboside, preferably no greater than 0.1 wt.% relative to the amount of reduced nicotinamide riboside, more preferably no greater than 0.01 wt.% relative to the amount of reduced nicotinamide riboside, most preferably no greater than 0.001 wt.% relative to the amount of reduced nicotinamide riboside.


Another aspect of the present invention is a method for increasing intracellular NAD+ in a mammal in need thereof, comprising administering to the mammal a composition consisting essentially of or consisting of reduced nicotinamide riboside in an amount effective to increase NAD+ biosynthesis. The method can promote the increase of intracellular levels of NAD+ in cells and tissues for improving cell and tissue survival and overall cell and tissue health, for example, in neuronal cells and tissues, especially in the brain.


Nicotinamide adenine dinucleotide (NAD+) is considered a coenzyme, and essential cofactor in cellular redox reactions to produce energy. It plays critical roles in energy metabolism, as the oxidation of NADH to NAD+ facilitates hydride-transfer, and consequently ATP generation through mitochondrial oxidative phosphorylation. It also acts as a degradation substrate for multiple enzymes (Canto, C. et al. 2015; Imai, S. et al. 2000; Chambon, P. et al. 1963; Lee, H.C. et al. 1991).


Mammalian organisms can synthesize NAD+ from four different sources. First, NAD+ can be obtained from tryptophan through the 10-step de novo pathway. Secondly, Nicotinic acid (NA) can also be transformed into NAD+ through the 3-step Preiss-Handler path, which converges with the de novo pathway. Thirdly, intracellular NAD+ salvage pathway from nicotinamide (NAM) constitutes the main path by which cells build NAD+, and occurs through a 2-step reaction in which NAM is first transformed into NAM-mononucleotide (NMN) via the catalytic activity of the NAM-phosphoribosyltransferase (NAMPT) and then converted to NAD+ via NMN adenylyltransferase (NMNAT) enzymes. Finally, Nicotinamide Riboside (NR) constitutes yet a fourth path to NAD+, characterized by the initial phosphorylation of NR into NMN by NR kinases (NRKs) (Breganowski, P. et al.; 2004).


An important difference between NR and NRH is that they go through different synthetic pathways to synthesis NAD+. For example, NRH does not use the NRK-1 enzyme pathway (J. Giroud-Gerbetant et al. 2019).


Five molecules previously have been known to act as direct extracellular NAD+ precursors: tryptophan, nicotinic acid (NA), nicotinamide (NAM), nicotinic acid riboside (NaR) and nicotinamide riboside (NR). The present invention discloses a new molecule that can act as an extracellular NAD+ precursor, reduced nicotinomide riboside (NRH). The reduction of the NR molecule to NRH confers it not only a much stronger capacity to increase intracellular NAD+ levels, but also a different selectivity in terms of its cellular use.


The present invention relates to NRH, a new molecule which can act as an NAD+ precursor. This reduced form of NR, which displays an unprecedented ability to increase NAD+ and has the advantage of being more potent and faster than nicotinamide riboside (NR). NRH utilizes a different pathway than NR to synthesize NAD+, which is NRK independent. NR and NRH use independent pathways in the biosynthesis of NAD+. The present invention demonstrates that NRH is protected against degradation in plasma and can be detected in circulation after oral administration. These advantages of the invention support its therapeutic efficacy.


The method comprises administering an effective amount of a composition consisting essentially of reduced nicotinamide riboside or consisting of reduced nicotinamide riboside to the individual.


In each of the compositions and methods disclosed herein, the composition is preferably a food product or beverage product, including food additives, food ingredients, functional foods, dietary supplements, medical foods, nutraceuticals, oral nutritional supplements (ONS) or food supplements.


The composition can be administered at least one day per week, preferably at least two days per week, more preferably at least three or four days per week (e.g., every other day), most preferably at least five days per week, six days per week, or seven days per week. The time period of administration can be at least one week, preferably at least one month, more preferably at least two months, most preferably at least three months, for example at least four months. In some embodiments, dosing is at least daily; for example, a subject may receive one or more doses daily, in an embodiment a plurality of doses per day. In some embodiments, the administration continues for the remaining life of the individual. In other embodiments, the administration occurs until no detectable symptoms of the medical condition remain. In specific embodiments, the administration occurs until a detectable improvement of at least one symptom occurs and, in further cases, continues to remain ameliorated.


The compositions disclosed herein may be administered to the subject enterally, e.g., orally, or parenterally. Non-limiting examples of parenteral administration include intravenously, intramuscularly, intraperitoneally, subcutaneously, intraarticularly, intrasynovially, intraocularly, intrathecally, topically, and inhalation. As such, non-limiting examples of the form of the composition include natural foods, processed foods, natural juices, concentrates and extracts, injectable solutions, microcapsules, nano-capsules, liposomes, plasters, inhalation forms, nose sprays, nosedrops, eyedrops, sublingual tablets, and sustained-release preparations.


The compositions disclosed herein can use any of a variety of formulations for therapeutic administration. More particularly, pharmaceutical compositions can comprise appropriate pharmaceutically acceptable carriers or diluents and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols. As such, administration of the composition can be achieved in various ways, including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, and intratracheal administration. The active agent may be systemic after administration or may be localized by the use of regional administration, intramural administration, or use of an implant that acts to retain the active dose at the site of implantation.


In pharmaceutical dosage forms, the compounds may be administered as their pharmaceutically acceptable salts. They may also be used in appropriate association with other pharmaceutically active compounds. The following methods and excipients are merely exemplary and are in no way limiting.


For oral preparations, the compounds can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose functional derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.


The compounds can be formulated into preparations for injections by dissolving, suspending or emulsifying them in an aqueous or non-aqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional, additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.


The compounds can be utilized in an aerosol formulation to be administered by inhalation. For example, the compounds can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like.


Furthermore, the compounds can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. The compounds can be administered rectally by a suppository. The suppository can include a vehicle such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at room temperature.


Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition. Similarly, unit dosage forms for injection or intravenous administration may comprise the compounds in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier, wherein each dosage unit, for example, mL or L, contains a predetermined amount of the composition containing one or more of the compounds.


Compositions intended for a non-human animal include food compositions to supply the necessary dietary requirements for an animal, animal treats (e.g., biscuits), and/or dietary supplements. The compositions may be a dry composition (e.g., kibble), semi-moist composition, wet composition, or any mixture thereof. In one embodiment, the composition is a dietary supplement such as a gravy, drinking water, beverage, yogurt, powder, granule, paste, suspension, chew, morsel, treat, snack, pellet, pill, capsule, tablet, or any other suitable delivery form. The dietary supplement can comprise a high concentration of the UFA and NORC, and B vitamins and antioxidants. This permits the supplement to be administered to the animal in small amounts, or in the alternative, can be diluted before administration to an animal. The dietary supplement may require admixing, or can be admixed with water or other diluent prior to administration to the animal.


REFERENCES

Bieganowski, P. and C. Brenner, 2004. Discoveries of nicotinamide riboside as a nutrient and conserved NRK genes establish a Preiss-Handler independent route to NAD+ in fungi and humans. Cell. 117(4): 495-502.


Callizot et al., 2013. Operational dissection of β-amyloid cytopathic effects on cultured neurons. J Neurosci Res. May;91(5):706-16.


Canto, C., K.J. Menzies, and J. Auwerx, 2015. NAD(+) Metabolism and the Control of Energy Homeostasis: A Balancing Act between Mitochondria and the Nucleus. Cell Metab. 22(1): 31-53.


Chambon, P., J.D. Weill, and P. Mandel, 1963. Nicotinamide mononucleotide activation of new DNA-dependent polyadenylic acid synthesizing nuclear enzyme. Biochem Biophys Res Commun. 1139-43.


Chen, L.K., et al. (2014). Sarcopenia in Asia: consensus report of the Asian Working Group for Sarcopenia. Journal of the American Medical Directors Association 15, 95-101.


Clark RV, Walker AC, O'Connor-Semmes RL, Leonard MS, Miller RR, Stimpson SA, Turner SM, Ravussin E, Cefalu WT, Hellerstein MK, Evans WJ (1985). Total body skeletal muscle mass: estimation by creatine (methyl-d3) dilution in humans. J Appl Physiol. Jun 15;116(12):1605-13.


Cruz-Jentoft, A.J., Baeyens, J.P., Bauer, J.M., Boirie, Y., Cederholm, T., Landi, F., Martin, F.C., Michel, J.P., Rolland, Y., Schneider, S.M., et al. (2010). Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 39, 412-423.


Fearon et al. (2011) Definition and classification of cancer cachexia: an international consensus. Lancet Oncology, 12, 489-495.


Giroud-Gerbetant, J. et al. (2019) A reduced form of nicotinamide riboside defines a new pathway for synthesis of NAD+ and acts as an orally bioavailable NAD+ precursor, Molecular Metabolism, Vol.30, pp.192-202.


Imai, S., C.M. Armstrong, M. Kaeberlein, and L. Guarente, 2000. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature. 403(6771): 795-800.


Lee, H.C. and R. Aarhus, 1991. ADP-ribosyl cyclase: an enzyme that cyclizes NAD+ into a calcium-mobilizing metabolite. Cell Regul. 2(3): 203-9.


Makarov, M. and M. Migaud, 2019. Syntheses and chemical properties of β-nicotinamide riboside and its analogues and derivatives. Beilstein J. Org. Chem. 15: 401-430.


Studenski SA, Peters KW, Alley DE, Cawthon PM, McLean RR, Harris TB, Ferrucci L, Guralnik JM, Fragala MS, Kenny AM, Kiel DP, Kritchevsky SB, Shardell MD, Dam TT, Vassileva MT (2014). The FNIH sarcopenia project: rationale, study description, conference recommendations, and final estimates. J Gerontol A Biol Sci Med Sci. 69(5), 547-558.


Stimpson SA, Leonard MS, Clifton LG, Poole JC, Turner SM, Shearer TW, Remlinger KS, Clark RV, Hellerstein MK, Evans WJ. (2013) Longitudinal changes in total body creatine pool size and skeletal muscle mass using the D3-creatine dilution method. J Cachexia Sarcopenia Muscle. Jun 25.


Ying (2007). NAD(+) and NADH in brain functions, brain diseases and brain aging. February 2007, Frontiers in Bioscience 12(5): 1863-88.





DESCRIPTION OF FIGURES


FIG. 1. Chemical structure of nicotinamide riboside in its oxidized (NR) and reduced (NRH) forms 1: 1-b-D-ribofuranosyl-3-pyridinecarboxamide salt 2: 1,4-dihydro-1-b-D-ribofuranosyl-3-pyridinecarboxamide 3: 1,2-dihydro-1-b-D-ribofuranosyl-3-pyridinecarboxamide 4: 1,6-dihydro-1-b-D-ribofuranosyl-3-pyridinecarboxamide X-: anion (e.g. triflate)



FIG. 2. Dose-response experiments revealed that NRH could significantly increase NAD+ better than NR Starting at levels at a concentration of 10 µM, NRH achieved similar increases in intracellular NAD+ levels to those reached with NR at 50-fold higher concentrations. NRH achieved maximal effects on NAD+ synthesis around the millimolar range, managing to increase intracellular NAD+ levels by more than 10-fold.



FIG. 3. NHR acts rapidly after 5 minutes from treatment. NRH actions were also extremely fast, as significant increases in NAD+ levels were observed within 5 minutes after NRH treatment. Peak levels of NAD+ were achieved between 45 minutes and 1 h after treatment.



FIG. 4. NRH leads to NAD+ biosynthesis through an adenosine kinase dependent path. AML12 cells were treated with an adenosine kinase inhibitor (5-IT; 10 mM) for 1 hour prior to NRH treatment at the doses indicated. Then, 1 hour later, acidic extracts were obtained to measure NAD+ levels. All values in the figure are expressed as mean +/- SEM of 3 independent experiments. * indicates statistical difference at p< 0.05 vs. the respective vehicle treated group.



FIG. 5. NRH is found intact in mice tissues after oral administration. 8 week-old C57BI/6NTac mice were orally gavaged with either saline (as vehicle), or NRH (250 mg/kg). After 2 hours, NAD+ levels in the brain were evaluated. All results are expressed as mean +/-SEM of n=4 mice per group as areas under the signal by LC-MS analysis corrected by total protein amount of tissue.



FIG. 6. NRH increases NAD+ levels in the brain after oral administration. 8 week-old C57BI/6NTac mice were orally gavaged with either saline (as vehicle) or NRH (500 mg/kg). After 1 hour, brain NAD+ was measured using commercial kits. All results are expressed as mean +/-SEM of n=4 mice per group, corrected by weight of the tissue.



FIG. 7. NRH protects against Amyloid β oligomer induced loss of neurite network. Primary rat cortical neurons were cultured as described in Callizot et al., 2013. On day 11 of culture, dihydronicotinamdie riboside (NRH; 0.01 mM) or brain derived neurothrophic factor (BDNF, 50 ng/mL, as positive control) were dissolved in the culture medium and pre-incubated with primary cortical neurons for the time indicated (24 hr or 6 hr), before exposure to Amyloid β-42 exposure as a way to model Alzheimer’s disease cellular damage. The Aβ1-42 preparation was done as described in Callizot et al., 2013. The Aβ1-42 preparation was added to a final concentration of 15 µmol/L (2.25 µmol/L oligomers, AβO) diluted in control medium in presence of compounds and let for 24 h. The cortical neurons were then fixed by a cold solution of ethanol (95%) and acetic acid (5%), permeabilized with saponin and incubated with an anti microtubule-associated-protein 2 (MAP-2), to stain cell bodies and neurites, allowing the study of the length of the neurite network. For each condition, 30 pictures (representative of the all well area) per well were taken using ImageXpress (Molecular Devices) with 20x magnification. Neurite length was then evaluated as % of control values. All values are expressed as mean +/-SEM from 5-6 independent wells. * indicates statistically significant difference vs. the amyloid b1-42 treated groups.





EXAMPLES
Example 1: Synthesis of the Reduced Form of Nicotinamide Riboside (NRH)

Reduced nicotinamide riboside (NRH) was obtained from NR (1) by reduction of pyridinium salts (for example, triflate) to dihydropyridines (1,2-, 1,4-, and 1,6-dihydropyridines) as shown below




embedded image




  • 1: 1-b-D-ribofuranosyl-3-pyridinecarboxamide salt

  • 2: 1,4-dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide

  • 3: 1,2-dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide

  • 4: 1,6-dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide

  • X-: anion (e.g. triflate)



Sodium borohydride (NaBH4) and sodium dithionite (Na2S2O4) were used as reducing agents for N-substituted pyridinium derivatives. Regioselectivity of reducing agents differ, leading to either only one dihydropyridine or a mixture of all 3 isomers in different proportions (2,3,4).


Dithionate reduction of pyridinium salts, carrying electron withdrawing substituents in positions 3 and 5, yielded almost exclusively 1,4-dihydropyridine products. The reduction was made in mild conditions (e.g. in aqueous sodium bicarbonate or potassium phosphate dibasic medium), due to instability of the reduced products in acidic media. To perform the reduction, hydroxyl groups in the ribofuranose moiety were protected with either benzyl or acetyl substituents. Deprotection was then be done by sodium hydroxide in methanol under ball mill conditions, after reduction.


Example 2: Measurement of NRH and Other NAD+ Related Metabolites in Biological Samples

Levels of NRH and other NAD-related metabolites in biological samples were obtained by using a cold liquid-liquid extraction using a mixture of methanol:water: chloroform in 5:3:5 (v/v), from which the polar phase was recovered for for hydrophilic interaction ultra-high performance liquid chromatography mass spectrometry (UHPLC-MS) analysis. The UHPLC consisted of a binary pump, a cooled autosampler, and a column oven (DIONEX Ultimate 3000 UHPLC+ Focused, Thermo Scientific), connected to a triple quadrupole spectrometer (TSQ Vantage, Thermo Scientific) equipped with a heated electrospray ionisation (H-ESI) source. Of each sample, 2 µL were injected into the analytical column (2.1 mm x 150 mm, 5 µm pore size, 200 Å HILICON iHILIC®-Fusion(P)), guarded by a pre-column (2.1 mm x 20 mm, 200 Å HILICON iHlLIC®-Fusion(P) Guard Kit) operating at 35° C. The mobile phase (10 mM ammonium acetate at pH 9, A, and acetonitrile, B) was pumped at 0.25 mL/min flow rate over a linear gradient of decreasing organic solvent (0.5-16 min, 90-25% B), followed by re-equilibration for a total run time of 30 min. The MS operated in positive mode at 3500 V with multiple reaction monitoring (MRM). The software Xcalibur v4.1.31.9 (Thermo Scientific) was used for instrument control, data acquisition and processing. Retention time and mass detection was confirmed by authentic standards.


Structure elucidation of the used NRH for biological studies was confirmed by nuclear magnetic resonance (NMR).


Example 3: NRH Is a Potent NAD+ Precursor

AML12 hepatocytes were treated with NRH, and it was observed that the ability of NRH to increase intracellular NAD+ was superior to that of NR.


Dose-response experiments revealed that NRH could significantly increase NAD+ levels at a concentration of 10 µM (FIG. 2). Even at such relatively low dose, NRH achieved similar increases in intracellular NAD+ levels to those reached with NR at 50-fold higher concentrations. NRH achieved maximal effects on NAD+ synthesis around the millimolar range, managing to increase intracellular NAD+ levels by more than 10-fold.


NRH actions were also extremely fast (FIG. 3), as significant increases in NAD+ levels were observed within 5 minutes after NRH treatment. Peak levels of NAD+ were achieved between 45 minutes and 1 h after treatment, as also occurred with NR.


The ability of NRH to potently increase NAD+ was tested as well in other cell type models. NRH treatment highly elevated NAD+ levels in C2C12 myotubes, INS1-cells and 3T3 fibroblasts, supporting the notion that NRH metabolism is widely conserved among different cell types.


Example 4: Pathway of NRH-induced NAD+ Synthesis

A path in which NRH would be converted to NMNH, then to NADH and this would be finally oxidized to NAD+. Accordingly, NRH and NMNH could be detected intracellularly 5 minutes after NRH, but not NR, treatment. Interestingly, NRH treatment also led to an increase in intracellular NR and NMN, greater than that triggered by NR itself, opening the possibility that NRH could synthesize NAD+ by being oxidized to NR, using then the canonical NRK/NMNAT path.


In order to understand the exact path by which NRH synthesizes NAD+, we initially evaluated whether NRH, could be transported into the cell by equilibrative nucleoside transporters (ENTs). Confirming this possibility, NRH largely lost its capacity as an extracellular NAD+ precursor in the presence of an agent blocking ENT-mediated transport, such as S-(4-nitrobenzyl)-6-thioinosine (NBTI). Nevertheless, a substantial action of NRH remained even after ENT blockage, suggesting that NRH might be able to enter the cell through additional transporters.


The action of NRH was also NAMPT-independent, based on experiments using FK866, a NAMPT inhibitor. If NRH led to NAD+ synthesis via the formation of NMNH, this hypothetical path would require the phosphorylation of NRH into NMNH. Given the essential and rate-limiting role of NRK1 in NR phosphorylation, we wondered whether the ability of NRH to boost NAD+ levels was NRK1 dependent. To answer this question, we evaluated NRH action in primary hepatocytes from either control or NRK1 knockout (NRK1 KO) mice. While after 1 hour of treatment NR failed to increase NAD+ levels in NRK1 KO derived primary hepatocytes, NRH action was not affected by NRK1 deficiency. These results indicate that NRH action is NRK1 independent. Further, they rule out the possibility that NRH-induced NAD+ transport is driven by NRH oxidation into NR.


Considering the molecular structure of NRH, we reasoned that an alternative nucleoside kinase could be responsible for the phosphorylation of NRH. Confirming this expectation, the adenosine kinase (AK) inhibitor 5-iodotubercidin (5-IT) fully ablated the action of NRH. The role of AK in NRH-mediated NAD+ synthesis was confirmed using a second, structurally different, AK inhibitor, ABT-702. Metabolomic analyses further confirmed that upon inhibition of AK, the generation of NMNH, NADH and NAD+ was fully blunted, even if NRH was effectively entering the cell. Interestingly, 5-IT treatment also prevented the formation of NR and NMN after NRH treatment.


This indicates that the occurrence of NR after NRH treatment cannot be attributed simply to direct NRH intracellular oxidation to NR. As a whole, these experiments depict adenosine kinase as the enzymatic activity catalyzing the conversion of NRH into NMNH, initiating this way the transformation into NAD+.


As a follow-up step, NMNAT enzymes could catalyze the transition from NMNH to NADH. Accordingly, the use of gallotannin as a NMNAT inhibitor largely compromised NAD+ synthesis after NRH treatment. Yet, part of the NRH action remained after gallotannin treatment when NRH was used at maximal doses. However, NRH action was totally blocked by gallotannin at submaximal doses, suggesting that the remaining effect at 0.5 mM could be attributed to incomplete inhibition of NMNAT activity by gallotannin. Altogether, these results indicate that adenosine kinase and NMNATs vertebrate the path by which NRH leads to NAD+ synthesis via NADH.


Example 5: NRH is Detectable in Circulation After IP Injection

NR degradation to NAM has been proposed as a limitation for its pharmacological efficacy. To evaluate whether NRH was also susceptible to degradation to NAM, we spiked NRH or NR in isolated mouse plasma. After 2 h of incubation, NR levels decayed in plasma, in parallel to an increase in NAM. In contrast, NAM was not generated from NRH, as its levels remained stable during the 2 h test. We also tested the stability of NRH in other matrixes. Given our previous experiments in cultured cells, we verified that NRH did not degrade to NAM in FBS supplemented media, as occurs with NR. Finally, we also certified NRH stability in water (pH=7, at room temperature) for 48 h.


The above results prompted us to test whether NRH could act as an effective NAD+ precursor in vivo. For this, we first intraperitoneally (IP) injected mice with either NR or NRH (500 mg/kg). After 1 h, both compounds increased NAD+ levels in liver, muscle and kidney. As expected, NAM levels were highly increased in circulation upon NR administration, while only a very mild increase was observed with NRH. Importantly, NRH was detectable in circulation after IP injection.


To our surprise, NR was detectable in circulation after NRH treatment at much higher levels than those detected after NR injection itself. Given that NRH incubation in isolated plasma did not lead to NR production, the appearance of NR might be consequent to intracellular production and release to circulation. Similarly, the residual appearance of NAM after NRH treatment might be explained by the degradation of released NR or by the release of intracellular NAM as a product of NAD+ degradation, as NRH did not significantly alter NAM levels when incubated in isolated plasma.


Example 6: NRH is Found Intact in Brain After Oral Administration

NRH is not only found in circulation but it was also found intact in brain 2 hours after gavage (FIG. 5). This indicates that oral administration of NRH allows for efficient biodistribution in brain tissues.


Example 7: NRH Increases NAD+ Levels in the Brain After Oral Administration

After intraperitoneal (I.P.) administration, NRH efficiently increases NAD+ levels in the brain, after 1 hour (FIG. 6). This indicates that I.P. administration of NRH allows for efficient biodistribution and bio-efficacy in the brain.


Example 8: NRH Protects Against Amyloid Β Oligomer Induced Loss of Neurite Network

Primary rat cortical neurons were cultured as described in Callizot et al., 2013. On day 11 of culture, dihydronicotinamdie riboside (NRH; 0.01 mM) or brain derived neurothrophic factor (BDNF, 50 ng/mL, as positive control) were dissolved in the culture medium and pre-incubated with primary cortical neurons for the time indicated (24 hr or 6 hr), before exposure to Amyloid β1-42 exposure as a way to model Alzheimer’s disease cellular damage. The Aβ1-42 preparation was done as described in Callizot et al., 2013. The Aβ1-42 preparation was added to a final concentration of 15 µmol/L (2.25 µmol/L oligomers, AβO) diluted in control medium in presence of compounds and let for 24 h. The cortical neurons were then fixed by a cold solution of ethanol (95%) and acetic acid (5%), permeabilized with saponin and incubated with an anti microtubule-associated-protein 2 (MAP-2), to stain cell bodies and neurites, allowing the study of the length of the neurite network. For each condition, 30 pictures (representative of the all well area) per well were taken using ImageXpress (Molecular Devices) with 20x magnification. Neurite length was then evaluated as % of control values. Results are shown in FIG. 7. This indicates that oral administration of NRH protects against Amyloid β-oligomer induced toxicity in terms of neurite length.

Claims
  • 1. A method of increasing intracellular NAD+ in a subject comprising delivering to the subject in need an effective unit dose form of reduced nicotinamide to prevent and/or treat neurological diseases or conditions.
  • 2. Method according to claim 1 wherein said reduced nicotinamide riboside is selected from the group consisting of: (i) 1,4-dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide;(ii) 1,2-dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide; and(iii) 1,6-dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide.
  • 3. Method according to claim 1 wherein the reduced nicotinamide riboside is preferably 1,4-dihydro-1-beta-D-ribofuranosyl-3-pyridinecarboxamide.
  • 4. comprising Method according to claim 1 wherein said dose is used to prevent and/or treat neurological diseases or conditions.
  • 5. comprising Method according to claim 1 wherein said dose is used to prevent and/or treat brain energy deficiency diseases or conditions.
  • 6. Method according to claim 1 wherein said dose consists essentially of reduced nicotinamide riboside without other NAD+ precursors to prevent and/or treat neurological diseases or conditions.
  • 7. Method according to claim 1 wherein said dose consists essentially of reduced nicotinamide riboside without other NAD+ precursors to prevent and/or treat brain energy deficiency diseases or conditions.
  • 8. Method according to claim 1 wherein the dose contains reduced nicotinamide riboside to maintain or improve neurological function in a subject.
  • 9. Method according to claim 1 to maintain or improve brain function in a subject.
  • 10. Method according to claim 9 wherein the brain function is motivational performance.
  • 11. Method according to claim 1 to enhance recovery of neurological function after injury or surgery.
  • 12. Method according to claim 1 wherein said dose is a nutritional composition selected from the group consisting of a: food or beverage product.
  • 13. Method according to claim 1 wherein the neurological disease and/or condition is the result of damage to the brain, spinal column or nerves caused by illness or injury.
  • 14. Method according to claim 1 wherein the neurological disease and/or condition is a brain energy deficiency disease and/or condition is selected from the group consisting of: migraine, memory disorder, age-related memory disorder, brain injury, neurorehabilitation, stroke and post-stroke, amyloid lateral sclerosis, multiple sclerosis, cognitive impairment, mild cognitive impairment (MCI), cognitive impairment post-intensive care, age-induced cognition impairment, Alzheimer’s disease, Parkinson’s disease, Huntingdon’s disease, inherited metabolic disorders such as glucose transporter type 1 deficiency syndrome and pyruvate dehydrogenase complex deficiency; bipolar disorder, schizophrenia, epilepsy, stress and motivational performance.
  • 15. Method to prevent and/or treat neurological diseases or conditions in a subject mammal, comprising delivering to the mammal in need of such treatment an effective amount of reduced nicotinamide riboside.
  • 16. Method for increasing intracellular NADH in a subject mammal, comprising delivering to the mammal in need of such treatment an effective amount of reduced nicotinamide riboside in an effective unit dose form to prevent and/or treat brain energy deficiency diseases or conditions.
  • 17. Method according to claim 15 comprising the steps of: i) providing the subject a composition consisting essentially of reduced nicotinamide riboside andii) administering the composition to said subject.
  • 18. Method according to claim 16. comprising the steps of: i) providing the subject a composition consisting essentially of reduced nicotinamide riboside andii) administering the composition to said subject.
  • 19. Method according to claim 15 wherein the subject is selected from the group consisting of: human, dog, cat, cow, horse, pig, and sheep.
  • 20. Method according to claim 19 wherein the subject is a human.
Priority Claims (1)
Number Date Country Kind
20161832.9 Mar 2020 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2021/055938 3/9/2021 WO