WO2013038740A1 - Method for screening for substance or factor capable of promoting muscle hypertrophy - Google Patents
Method for screening for substance or factor capable of promoting muscle hypertrophy Download PDFInfo
- Publication number
- WO2013038740A1 WO2013038740A1 PCT/JP2012/057128 JP2012057128W WO2013038740A1 WO 2013038740 A1 WO2013038740 A1 WO 2013038740A1 JP 2012057128 W JP2012057128 W JP 2012057128W WO 2013038740 A1 WO2013038740 A1 WO 2013038740A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- muscle
- factor
- peroxynitrite
- muscle hypertrophy
- superoxide
- 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
Links
Images
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/5044—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
- G01N33/5061—Muscle cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
- A61P21/04—Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
Definitions
- the present invention relates to a method for screening a substance or factor that promotes muscle hypertrophy and a muscle hypertrophy promoter.
- Non-patent Document 1 Skeletal muscle weight is controlled by the balance between protein synthesis and degradation (Non-patent Document 1).
- Muscle atrophy is not only immobilized by space flight under microgravity (Non-Patent Document 2) or tail suspension (Non-Patent Document 3), but also casheia (Non-Patent Document 4), aging (Sarkopair) (Non-Patent Document) 5), also caused by steroid administration (Non-patent Document 6).
- Non-patent Document 7 amyotrophic lateral sclerosis
- Neurogenic muscle atrophy or molecular pathology in the sense accompanying muscle atrophy
- Non-patent Document 8 amyotrophic lateral sclerosis
- nNOS neuronal nitric oxide synthase
- Non-patent Document 3 NNOS released from the muscle cell membrane during mouse tail suspension moves to the cytoplasm, produces nitric oxide (NO), inhibits phosphorylation of Forkheadkbox O (Foxo), and activates expression of E3 ubiquitin ligase Caused muscle atrophy.
- nNOS was involved in recovery from muscle wasting during reloading. This suggests that nNOS controls not only muscle atrophy but also muscle hypertrophy.
- the present invention aims to clarify the function of nNOS during muscle hypertrophy and to establish a new treatment method for muscle atrophy based on its molecular basis, that is, a method and means for promoting muscle hypertrophy. To do.
- the present inventors have found that peroxynitrite produced by NO and superoxide controls intracellular calcium concentration via TRP channels, thereby causing muscle hypertrophy. It was found that it was promoted, and it was found that muscle hypertrophy can be promoted by increasing the concentration of superoxide and / or peroxynitrite and / or calcium in muscle cells, leading to the completion of the present invention. It was.
- the present invention includes the following [1] to [9].
- [1] A screening method for substances or factors that promote muscle hypertrophy, (A) measuring the concentration of superoxide and / or peroxynitrite and / or calcium in muscle cells obtained from an animal treated with a test substance or factor; (B) A method comprising identifying a test substance or factor as a candidate substance or factor that promotes muscle hypertrophy based on the result of (a).
- [2] A screening method for substances or factors that promote muscle hypertrophy, (A) treating muscle cells with a test substance or factor; (B) measuring the concentration of superoxide and / or peroxynitrite and / or calcium in the muscle cells; (C) A method comprising identifying a test substance or factor as a candidate substance or factor that promotes muscle hypertrophy based on the result of (b). [3] The method according to [1] or [2], further comprising a step of measuring superoxide and / or peroxynitrite and / or calcium concentration in muscle cells before treatment with the test substance or factor. .
- the test substance or factor is a substance or factor that promotes muscle hypertrophy.
- a muscle hypertrophy promoter comprising at least one drug selected from the group consisting of a superoxide donor, a peroxynitrite donor, and a TRP channel agonist.
- a method for promoting muscle hypertrophy comprising administering to a subject an effective amount of at least one drug selected from the group consisting of a superoxide donor, a peroxynitrite donor, and a TRP channel agonist.
- the screening method according to the present invention can identify substances or factors that promote muscle hypertrophy and is useful for the development of therapeutic methods such as muscle atrophy.
- the muscle hypertrophy promoter according to the present invention can effectively promote muscle hypertrophy and is useful for prevention or treatment of muscle atrophy.
- the present invention provides a method and a muscle hypertrophy promoter for screening for substances and factors that promote muscle hypertrophy based on the involvement of superoxide, peroxynitrite and intracellular calcium in muscle hypertrophy.
- the present inventor performed hindlimb cooperative muscle excision in nNOS wild-type and deficient mice to induce compensatory muscle hypertrophy due to overload.
- the wild type increased the muscle weight by about 40% compared to the control group, but the deficient type increased only about 20%.
- NOS activity was measured.
- nNOS was activated 3 minutes after overloading and decreased to a steady level after 1 hour.
- pharmacological inhibition of nNOS activity it was found that NO produced during 1 hour of overload promoted muscle hypertrophy.
- vasodilators such as 8Br-cGMP cannot restore the defective phenotype, and NO produced by nNOS is mediated by vasodilation controlled by the classical NO-cGMP pathway. It was suggested that muscle hypertrophy was promoted. Therefore, it is a cGMP-independent pathway, and it is thought that peroxynitrite, which is a reaction product of NO and superoxide, may promote muscle hypertrophy. Superoxide and peroxynitrite immediately after overload An experiment was conducted to clarify the function of (Example 2).
- BAPTA-AM and EGTA calcium chelators
- nNOS-deficient phenotype was restored by NO donor and peroxynitrite donor, but the effect was completely suppressed by co-administration with BAPTA-AM and BTP2. Furthermore, administration of a TRP channel agonist (Hyp9, olvanil) restored the nNOS-deficient phenotype and increased muscle weight. Furthermore, it was found that peroxynitrite is involved in the increase of calcium concentration in myoblasts, specifically, inducing calcium release in the sarcoplasmic reticulum.
- a substance or factor that promotes or inhibits muscle hypertrophy is identified based on the concentration of superoxide and / or peroxynitrite and / or calcium in muscle cells. That is, by treating an animal or muscle cell with a test substance or factor and measuring the concentration of superoxide and / or peroxynitrite and / or calcium in the animal or muscle cell, the test substance or factor becomes superoxide. And / or whether it affects peroxynitrite and / or calcium concentrations, ie muscle hypertrophy.
- muscle hypertrophy refers to an increase in muscle weight due to an increase in the weight of a single muscle fiber or cross-sectional area accompanying an increase in the amount of endogenous protein
- promotes muscle hypertrophy By promoting the increase in amount, it means promoting the increase in muscle weight due to the increase in single muscle fiber weight or cross-sectional area.
- the screening method in the screening method according to the present invention (hereinafter, also referred to as “the screening method”), muscle cells obtained from an animal treated with a test substance or factor are prepared, or muscle cells are treated with the test substance or factor. Then, the superoxide and / or peroxynitrite and / or calcium concentration in the muscle cells is measured. Preferably, prior to treatment with a test substance or factor, superoxide and / or peroxynitrite and / or calcium concentrations in muscle cells or muscle cells obtained from animals are measured.
- the target animal is not particularly limited as long as it has muscles, and mammals such as humans, primates (monkeys, chimpanzees, etc.), livestock animals (cattle, horses, pigs, etc.), pets ( Dogs, cats, etc.), laboratory animals (mouse, rats, monkeys, etc.), amphibians, reptiles and birds. Further, the animal may be a normal animal, an muscularly atrophic animal, an nNOS-deficient animal, or an animal that has developed muscular dystrophy. In general, after the effectiveness of a test substance or factor is confirmed at the cellular level, the effectiveness is evaluated in a laboratory animal, and further in a human, for example, by a clinical test.
- test substance or factor that is the subject of this screening method is not particularly limited.
- the test substance or factor can be any substance, specifically a naturally occurring molecule such as an amino acid, peptide, oligopeptide, polypeptide, protein, nucleic acid, lipid, carbohydrate (such as sugar), steroid, glycopeptide Synthetic analogs or derivatives of naturally occurring molecules, such as peptidomimetics, nucleic acid molecules (aptamers, antisense nucleic acids, double-stranded RNA (RNAi), etc.), etc .; non-naturally occurring molecules, such as And low molecular organic compounds (inorganic and organic compound libraries, combinatorial libraries, etc.) produced using combinatorial chemistry techniques and the like; and mixtures thereof.
- the test substance or factor may be a single substance, a complex composed of a plurality of substances, a transcription factor, or the like.
- the test substance or factor may be an environmental factor such as radiation, ultraviolet rays, carbon concentration, or temperature.
- test substance or factor a single test substance or factor may be tested independently, or a mixture of several candidate test substances or factors (including a library or the like) may be tested.
- the library containing a plurality of test substances or factors include a synthetic compound library (such as a combinatorial library) and a peptide library (such as a combinatorial library).
- the treatment conditions such as the treatment amount, treatment period, treatment route, etc. vary depending on the type of test substance or factor, but those skilled in the art can easily determine it. it can.
- the route of administration is intramuscular injection, oral administration, intravenous injection, intraperitoneal injection, transdermal administration, subcutaneous injection, depending on the type of test substance and the type of animal used.
- Such administration forms can be appropriately used.
- Muscle cells can be collected by methods known in the art. Specifically, for example, after chopping skeletal muscle and transferring it to a 50 ml conical bottom tube, 4 ml of Dispase2 (2.4 IU / ml) -Collagenase XI (0.2%) solution per 1 g of muscle weight is added, Incubate at 37 ° C for 45-60 minutes, pipetting with a pipette every 15 minutes. Thereafter, the tissue piece is crushed by passing several times through an 18 G injection needle, and the supernatant is collected.
- the precipitated cells are suspended in 25 ml of growth medium, transferred to an uncoated 15 cm culture dish, added with bFGF, and cultured for 90 minutes at 37 ° C., 5% CO 2 concentration, and the supernatant containing unattached cells is collected. Using this supernatant, wash the bottom of the culture dish, change the direction 180 degrees, incubate again at 37 ° C, 5% CO 2 concentration for 90 minutes, and collect the supernatant. Transfer the collected supernatant to a 15 cm collagen-coated culture dish, add bFGF, and culture overnight at 37 ° C. and 5% CO 2 concentration. The next day, if the myoblasts are more than 30-40% confluent, passaging, and if less, change the medium. In order to prevent myogenic differentiation, subculture or medium exchange is performed every day thereafter. Alternatively, commercially available muscle cells or publicly available muscle cells can be used.
- the contact condition varies depending on the type of the substance or factor, but can be easily determined by those skilled in the art.
- such contact can be achieved by culturing myocytes in a medium supplemented with a test substance, immersing the myocytes in a solution containing the test substance, and laminating the test substance on the muscle cells.
- it can be performed by culturing myocytes in the presence of a test factor.
- the effect and effectiveness of the test substance or factor can be examined under several conditions. Such conditions include time or duration, amount (large or small), number of times, etc. of treatment with the test substance or factor.
- a plurality of doses can be set by preparing a dilution series of the test substance.
- the treatment period of the test substance or factor can also be set as appropriate.
- the treatment can be performed over a period of 1 day to several weeks, months, and years.
- test substances and / or factors may be used in combination.
- the concentration of superoxide and / or peroxynitrite and / or calcium in the muscle cell obtained from the animal or the muscle cell is appropriately determined. taking measurement. For example, immediately after treatment, 30 minutes, 1 hour, 3 hours, 5 hours, 10 hours, 15 hours, 20 hours, 24 hours (1 day), 2-10 days, 10-20 Measurements are taken after 20-30 days, 1-6 months later.
- the measurement of superoxide can be performed by a method known in the art, for example, by using lucigenin that emits light by reacting with superoxide, and measuring the emission intensity.
- Peroxynitrite can be measured by a method known in the art.
- the peroxynitrite is measured by measuring the fluorescence intensity of a fluorescent substance emitted by reacting peroxynitrite with an active oxygen species or an active nitrogen species. This can be done by calculating the difference from the control group treated with the nitric acid scavenger.
- the calcium concentration in muscle cells can also be measured by a method known in the art.
- the fluorescence intensity is measured using Fluo-4 that emits fluorescence when bound to calcium. Can be performed.
- test substance or factor that increases or decreases the superoxide and / or peroxynitrite and / or calcium concentration relative to the control after measuring the superoxide and / or peroxynitrite and / or calcium concentration in muscle cells Selected as a substance or factor that promotes or inhibits muscle hypertrophy.
- animals or muscle cells that have not been treated with the test substance or factor can be used.
- test substance or factor that has been treated with a test substance or factor and that has increased the superoxide and / or peroxynitrite and / or calcium concentration in the myocyte is obtained.
- test substance or factor Selected and then as a secondary screen, animals were treated with the selected test substance or factor and the concentrations of superoxide and / or peroxynitrite and / or calcium in the muscle cells of the animals were measured to show increased concentrations
- a test substance or factor may be selected.
- the selected test substance or factor is administered to an experimental animal to determine whether the test substance or factor promotes muscle hypertrophy in the experimental animal.
- an experimental animal a model animal, preferably a mouse, in which muscle atrophy has been induced by hindlimb suspension, denervation, dexamethasone administration, or the like can be used.
- Whether or not a test substance or factor promotes muscle hypertrophy in an experimental animal depends on the type of experimental animal and the like, but can be appropriately determined by those skilled in the art by methods known in the art. For example, muscle tissue can be collected from an animal and the muscle weight can be measured.
- a candidate substance or factor that promotes muscle hypertrophy can be identified, and further, the effectiveness of the substance or factor that promotes muscle hypertrophy can be confirmed.
- the muscle hypertrophy promoter according to the present invention contains a substance or factor capable of increasing the superoxide and / or peroxynitrite and / or calcium concentration.
- the muscle hypertrophy promoter according to the present invention includes at least one drug selected from the group consisting of a superoxide donor, a peroxynitrite donor, and a TRP channel agonist.
- the muscle hypertrophy promoter according to the present invention is useful for the prevention or treatment of diseases or disorders such as muscle atrophy, especially for the treatment of patients with severe muscle atrophy and difficulty in exercise therapy such as rehabilitation and for the increase in muscle mass of bedridden patients. It is.
- any drug can be used as long as it is a drug known to donate superoxide in this technical field.
- any drug can be used as long as it is a drug known to donate peroxynitrite in this technical field.
- SIN-1, molsidomine, etc. are included and all are commercially available.
- TRP channel agonist any drug can be used as long as it is a drug known in the art to have agonist activity with respect to the TRP channel.
- Hyp9, olvanil and the like are included, and commercially available products can be used.
- the muscle hypertrophy promoter may contain, as an active ingredient, one superoxide donor and / or a peroxynitrite donor and / or a TRP channel agonist, or two or more superoxide donors And / or a combination of a peroxynitrite donating agent and / or a TRP channel agonist.
- this muscle hypertrophy promoter contains as an active ingredient at least one drug selected from the group consisting of a superoxide donor, a peroxynitrite donor, and a TRP channel agonist, it exerts the above-described muscle hypertrophy promoter action. .
- the muscle hypertrophy promoter may contain a pharmaceutically acceptable carrier or additive in addition to a superoxide donor and / or a peroxynitrite donor and / or a TRP channel agonist as active ingredients.
- a pharmaceutically acceptable carrier or additive include water, pharmaceutically acceptable organic solvents, collagen, polyvinyl alcohol, polyvinyl pyrrolidone, carboxyvinyl polymer, sodium alginate, water-soluble dextran, sodium carboxymethyl starch, pectin, xanthan gum, Examples include gum arabic, casein, gelatin, agar, glycerin, propylene glycol, polyethylene glycol, petrolatum, paraffin, stearyl alcohol, stearic acid, human serum albumin, mannitol, sorbitol, and lactose.
- the additive to be used is appropriately or in combination selected from the above depending on the dosage form.
- tablets, capsules (hard capsules, soft capsules, microcapsules, etc.), granules, powders, pills, troches, liquids for internal use, liquids, suspensions, Any of emulsion, syrup, etc. may be used, and it may be a dry preparation re-dissolved when used.
- intravenous injection including infusion
- intramuscular injection intraperitoneal injection
- subcutaneous injection eg, solution, emulsion, suspension
- ointment Preparations such as pills, creams, suppositories, cataplasms, inhalants, liniments, aerosols and other external preparations can be selected.
- injections they are provided in unit dose ampoules or multi-dose containers Is done.
- compositions are excipients, extenders, binders, wetting agents, disintegrating agents, lubricants, surfactants, dispersants, buffering agents, pH adjusting agents, preservatives, solubilizers commonly used in medicine.
- Agents, preservatives, flavoring agents, absorption promoters, soothing agents, stabilizers, tonicity agents and the like can be appropriately selected and produced by conventional methods.
- the superoxide donor and / or peroxynitrite donor and / or TRP channel agonist to be added to the muscle hypertrophy promoter varies depending on the type of active ingredient, application, dosage form, route of administration, etc. 0.01 to 90% by weight, preferably 1 to 50% by weight.
- the effective amount (dose or intake) of the muscle hypertrophy promoter varies depending on the type of active ingredient contained in the agent, the age and weight of the subject, the administration route, and the number of administrations, and can be varied over a wide range.
- the effective amount of a superoxide donor and / or peroxynitrite donor and / or TRP channel agonist per day for an adult is usually about 1 week to about 1 year, preferably once or several times a day. Can be administered for about 1 month to about 12 months.
- This muscle hypertrophy promoter does not particularly limit the target (subject) to be used.
- it can be administered to or ingested by subjects such as humans, domestic animals (such as cows), pets (such as dogs and cats), and laboratory animals (such as monkeys).
- This muscle hypertrophy promoter can be combined with other muscle hypertrophy promoters known in the art or methods effective for promoting muscle hypertrophy. For example, it is combined with administration of protein that is a nutrient necessary for muscle hypertrophy, administration of hormones (growth hormone, etc.) that assimilate the nutrient to the muscle, load on the muscle (eg, mild exercise, strength training, pressure training), etc. be able to.
- hormones growth hormone, etc.
- the present muscle hypertrophy promoter is not limited to the use as a pharmaceutical composition, and may be blended in other products such as food or feed.
- Food and “feed” refer to natural products containing one or more nutrients and processed products thereof, including all food and drink.
- a food or feed containing the present muscle hypertrophy promoter is useful as a health supplement product for promoting muscle hypertrophy.
- solid food includes bread dough; dough for baked confectionery such as rice crackers, biscuits and cookies; noodles such as buckwheat and udon; fish products such as kamaboko and chikuwa; livestock products such as ham and sausage; powdered milk and the like It is done.
- jelly-like food include fruit jelly and coffee jelly.
- liquid foods include beverages such as soft drinks and fruit beverages (tea, coffee, tea, fermented milk, lactic acid bacteria beverages, etc.), seasonings (mayonnaise, dressings, seasoning seasonings, etc.).
- capsule foods include hard capsules and soft capsules.
- the amount added is 0.01 to 90% by weight of the superoxide donor and / or peroxynitrite donor and / or TRP channel agonist with respect to the whole food. It can mix
- the intake that can be expected to be effective is appropriately determined according to the individual case, taking into account the age, weight, sex, and degree of symptoms.
- the number of intakes can be divided into several times a day, in which case the amount can be divided according to the number of times. In addition, it can be taken continuously over a long period of time.
- Example 1 In this example, in order to clarify the contribution of nNOS in muscle hypertrophy, cooperative muscle excision was performed on nNOS wild-type and deficient mice to induce compensatory muscle hypertrophy due to overload. Subjects include Jackson Laboratories (Wilmington, NNOS-deficient mice purchased from MA) and C57BL / 6 mice purchased from CLEA Japan (Japan) were used.
- Compensatory muscle hypertrophy induction and drug administration by synergistic muscle resection were performed as follows. That is, the gastrocnemius and soleus tendons of 12-week-old mice were excised under anesthesia (Adams, GR and F. Haddad, J Appl Physiol, 1996. 81 (6): 2509-16). The control group underwent sham surgery to cut the skin. After surgery, 1 mg / ml ampicillin was added to drinking water. The NOS inhibitor L-NAME (1 mg / ml, Sigma-Aldrich) was administered as drinking water and the drinking water was changed daily.
- nNOS inhibitor 7-NI 50 mg / kg, Calbiochem
- NOX inhibitor DPI 2 mg / kg, Sigma-Aldrich
- apocynin (10 mg / kg, Sigma-Aldrich)
- tadalafil (10 mg / kg, Toronto Research Chemicals)
- sildenafil (10 mg / kg, Sigma-Aldrich)
- 8-Br-cGMP 10 mg / kg, Calbiochem
- Rotenone (5 mg / kg, Sigma-Aldrich), allopurinol (10 mg / kg, Cayman), uric acid (200 mg / kg Sigma-Aldrich; Example 2), FeTPPS (30 mg / kg, Calbiochem; Example) 2), ebselen (30 mg / kg, Sigma-Aldrich; Example 2), molsidomine (50 mg / kg, Sigma-Aldrich; Example 2), SIN-1 (10 mg / kg
- BAPTA-AM 50 ⁇ M, Calbiochem; Example 3
- EGTA 1 mM, Dojindo; Example 3
- dantrolene 10 ⁇ M, Sigma-Aldrich; Example 3
- nifedipine 10 ⁇ M, Sigma-Aldrich; Example 3)
- BTP2 10 ⁇ M, Calbiochem; Example 3
- Gd 3+ 50 ⁇ m, Sigma-Aldrich; Example 3
- GsMTx-4 10 ⁇ M, Wako; Example 3
- Hyp9 10 ⁇ M, Sigma-Aldrich; Example 3)
- olvanil 10 ⁇ M, Sigma-Aldrich; Example 3) were intramuscularly injected 30 minutes before the start of overload.
- the plantar muscle was frozen and fixed, sliced to 8 ⁇ m, and stained with hematoxylin and eosin.
- sample buffer (0.1% Triton X-100, 50 mM HEPES (pH7.4), 4 mM EGTA, 10 mM EDTA, 15 mM Na 4 P 2 O 7 , 100 mM glycerophosphate, 25 mM NaF, 5 mM Na 2 VO 4 , and complete protease inhibitor cocktail (Roche)) and centrifuged (15,000 g, 10 minutes), and the supernatant was collected.
- sample buffer 0.1% Triton X-100, 50 mM HEPES (pH7.4), 4 mM EGTA, 10 mM EDTA, 15 mM Na 4 P 2 O 7 , 100 mM glycerophosphate, 25 mM NaF, 5 mM Na 2 VO 4 , and complete protease inhibitor cocktail (Roche)
- centrifuged (15,000 g, 10 minutes), and the supernatant was collected.
- sample loading buffer (30% glycerol, 5% 2-mercaptoethanol, 2.3% SDS, 62.5 mM Tris-HCl ( pH 6.8), 0.05% bromophenol blue), heat-denatured at 60 ° C. for 15 minutes, and 30 ⁇ g was subjected to Western blotting.
- the PVDF transfer membrane was blocked with Tris-buffered saline (TBS) + 5% skim milk (snow mark), and incubated for 16 hours at 4 ° C. using the primary antibody.
- TBS Tris-buffered saline
- Primary antibodies include Akt (# 9272, Cell Signaling Technology), p-Akt (Ser473) (# 9271, Cell Signaling Technology), p70S6K (# 9202, Cell Signaling Technology), and p-p70S6K (Thr389) (# 9205 , Cell Signaling Technology).
- the activity was measured by measuring the amount of citrulline which is a by-product during NO production.
- the thinned plantar muscle is homogenized with 10 times the amount of measurement buffer (25 mM Tris-HCl (pH 7.4), 1 mM EDTA, 1 mM EGTA, and 0.1 mM NaCl) and centrifuged (1,000 g, The supernatant was collected after 10 minutes).
- reaction stop buffer (20 mM HEPES (pH 5.5) containing 2 mM EDTA) was added, and then mixed with AG50WX-8 columns (Na + form; Dowex) to recover unreacted [ 3 H] arginine Thereafter, the amount of [ 3 H] citrulline was measured with a liquid scintillation counter.
- the student-t test was used to compare the data between the two groups. For comparison between multiple groups, ANOVA test was performed and then multiple group test by Tukey's method was performed. Data are shown using mean ⁇ standard error, and p ⁇ 0.05 was determined to be significant.
- the wild type increased the muscle weight by about 40% compared to the control group, but the deficient type increased only about 20% (Fig. 1).
- a and B In order to clarify the contribution of NO in muscle hypertrophy, the NOS inhibitor L-NAME was administered as described above. In the group administered L-NAME from 3 days before to 7 days after surgery, and from 3 days before surgery to 1 day after surgery, muscle weight was significantly reduced on the 7th day after surgery, but in the group administered after 1 day after surgery, No effect was seen (Figure 2). To examine whether NO production was due to nNOS, 7-NI, an nNOS-specific inhibitor, was administered 30 minutes before or 60 minutes after overload.
- phosphorylation of p70S6K threonine 389 (Zoncu, R., A. Efeyan, and DM Sabatini, Nat Rev Mol Cell Biol, 2011. 12 (1): 21-: 35) increased, mTOR was activated, and the protein synthesis pathway was activated, but the activation was not observed in the defective type (A and B in FIG. 5).
- nNOS activated immediately after overload promotes muscle hypertrophy through activation of protein synthesis pathway controlled by mTOR.
- vasodilators tadalafil, sildenafil, and 8Br-cGMP which activates the cGMP pathway
- Example 2 In this example, the contribution of mitochondrial respiratory chain, xanthine oxidase, and NADPH oxidase (NOX), which are main supply molecules of superoxide in skeletal muscle, to muscle hypertrophy was examined.
- Superoxide production was measured by measuring luminescence by reaction between superoxide and lucigenin. After freezing, homogenize sliced plantar muscle with measurement buffer (150 mM NaCl, 50 mM Tris-HCl (pH 7.4), 25 mM EGTA, 25 mM EDTA, and complete protease inhibitor cocktail (Roche)), The supernatant was collected after 1,000 g for 10 minutes. The supernatant was mixed with measurement buffer (100 mM KH 2 PO 4 , (pH 7.0) and 10 ⁇ M lucigenin (Sigma-Aldrich)). 200 ⁇ M NADPH was added to the reaction solution immediately before the measurement and measured with a liquid scintillation counter. As a result, production of superoxide did not increase immediately after overload (FIG. 7).
- NOX1, NOX2, NOX3 and NOX4 in the plantar muscle was examined by RT-PCR using a primer set having the following sequence.
- NOX2 and NOX4 are expressed in the plantar muscles (FIG. 8), and NOX4 is a constantly activated enzyme (Bedard, K. and KH Krause, Physiol Rev, 2007. 87 (1): 245-313) It has been reported that NOX4 promotes muscle hypertrophy.
- Example 3 Peroxynitrite can control intracellular calcium concentration (Trebak, M., et al., Antioxid Redox Signal, 2010. 12 (5): 657-74). It was thought that mTOR is controlled via control and promotes muscle hypertrophy. In order to clarify the relationship between intracellular calcium concentration and mTOR activation, intramuscular injection of thapsigargin that inhibits calcium uptake by sarcoplasmic reticulum and raises intracellular calcium concentration resulted in activation of mTOR (A and B in FIG. 12).
- BAPTA-AM and EGTA which are calcium chelating agents, were injected intramuscularly.
- the muscle weight on the 7th day after the operation was significantly suppressed (FIG. 13).
- inhibitors of ryanodine receptor, L-type calcium channel, and TRP channel (dantrolene, nifedipine, BTP2), which are typical calcium channels in skeletal muscle, were injected intramuscularly.
- the muscle weight on the 7th day after the operation was significantly suppressed by the TRP channel inhibitor BTP2, but not by dantrolene and nifedipine (FIG. 13).
- the nNOS deficient phenotype was recovered by NO donor (SNAP) and peroxynitrite donor (SIN-1), but the effect was suppressed by co-administration with BAPTA-AM and BTP2 (FIG. 15).
- the deficient phenotypes are TRP channel agonists, ie, TRPC6 channel agonists Hyp9 (Leuner, K., et al., Mol Pharmacol, 2010. 77 (3): 368-77) and TRPV1 channel agonists It was recovered by administration of olvanil (FIG. 16).
- C2C12 that induced myogenic differentiation was cultured in PSS solution (140 mM NaCl, 5 mM KCl, 2.5 mM CaCl 2 , 1 mM MgCl 2 , 10 mM HEPES, 10 mM glucose pH 7.0) for 6 hours or more, and then 8 ⁇ M Fluo-4 was added. And incubated at room temperature for 30 minutes. After removing excess Fluo-4 and culturing at 37 ° C. for 5 minutes, changes in fluorescence intensity due to SIN-1 (DOJINDO) were measured every 3 seconds using an inverted fluorescence microscope (Olympus).
- PSS solution 140 mM NaCl, 5 mM KCl, 2.5 mM CaCl 2 , 1 mM MgCl 2 , 10 mM HEPES, 10 mM glucose pH 7.0
- SIN-1 When extracellular calcium was removed, SIN-1 was added to the cells using 0 Ca2 + solution (140 mM NaCl, 5 mM KCl, 1 mM MgCl2, 10 mM HEPES, 10 mM glucose, 2 mM EGTA pH 7.0).
- FeTPPS calbiochem
- thapsigargin a peroxynitrite scavenger
- the increase in intracellular calcium concentration occurs due to the uptake of extracellular calcium or the release of calcium stored in the intracellular sarcoplasmic reticulum.
- Extracellular calcium was removed (0Ca2 +), or calcium in the sarcoplasmic reticulum was depleted by thapsigargin.
- SIN-1 the increase in intracellular calcium concentration by SIN-1 was suppressed by depletion of sarcoplasmic reticulum calcium (FIGS. 19A and 19B).
- Sequence numbers 1 to 10 Artificial sequence (synthetic oligonucleotide)
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Cell Biology (AREA)
- Chemical & Material Sciences (AREA)
- Biomedical Technology (AREA)
- General Health & Medical Sciences (AREA)
- Urology & Nephrology (AREA)
- Medicinal Chemistry (AREA)
- Hematology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Molecular Biology (AREA)
- Analytical Chemistry (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Food Science & Technology (AREA)
- Biotechnology (AREA)
- Physics & Mathematics (AREA)
- Tropical Medicine & Parasitology (AREA)
- Biochemistry (AREA)
- Toxicology (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Neurology (AREA)
- Microbiology (AREA)
- Physical Education & Sports Medicine (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Description
本発明は、筋肥大を促進する物質又は因子のスクリーニング方法、及び筋肥大促進剤に関する。 The present invention relates to a method for screening a substance or factor that promotes muscle hypertrophy and a muscle hypertrophy promoter.
骨格筋重量はタンパク質合成と分解のバランスによって制御されている(非特許文献1)。ギプス固定やベッドレストなどの筋不活動状態においては、タンパク質合成の抑制及びタンパク質分解の促進により、筋タンパク質含有量が低下し、結果として筋量が減少し、筋が萎縮する(廃用性筋萎縮)。筋萎縮は、微小重力下の宇宙飛行(非特許文献2)や、尾部懸垂(非特許文献3)などによる不動化だけでなく、カヘキシア(非特許文献4)、老化(サルコペア)(非特許文献5)、ステロイド投与(非特許文献6)などによっても生ずる。 Skeletal muscle weight is controlled by the balance between protein synthesis and degradation (Non-patent Document 1). In muscle inactive states such as cast casts and bed rest, the protein content is decreased due to the inhibition of protein synthesis and the promotion of proteolysis, resulting in a decrease in muscle mass and muscle atrophy (disused muscle atrophy). Muscle atrophy is not only immobilized by space flight under microgravity (Non-Patent Document 2) or tail suspension (Non-Patent Document 3), but also casheia (Non-Patent Document 4), aging (Sarkopair) (Non-Patent Document) 5), also caused by steroid administration (Non-patent Document 6).
筋萎縮に対する治療法の開発は、高齢化社会を迎えた日本において非常に大きな課題の一つである。筋肥大を誘導、あるいは筋萎縮を軽減させる手段を得ることができれば、神経原性の筋萎縮を生じる筋萎縮性側索硬化症(非特許文献7)、また筋萎縮を伴う意味では分子病態が共通する可能性のある筋ジストロフィー(非特許文献8)に対する効果的な手段の開発にも結びつくことが期待される。 Developing treatments for muscle atrophy is one of the major challenges in Japan, which has faced an aging society. If a means for inducing muscle hypertrophy or reducing muscle atrophy can be obtained, amyotrophic lateral sclerosis (Non-patent Document 7) that causes neurogenic muscle atrophy, or molecular pathology in the sense accompanying muscle atrophy, It is expected to lead to the development of effective means for muscular dystrophy (Non-patent Document 8) that may be common.
タンパク質合成を活性化させることで筋肥大を誘導することは、筋萎縮に対する治療として期待が持たれる。重度の筋萎縮を伴う患者や寝たきりの高齢者に対し、一般的な運動療法を用いることは非常に難しく、薬物治療が望まれる。これまでPI3K/Akt経路を活性化させるIgf-1(非特許文献9)を投与することでタンパク質合成を活性化させる研究が行われてきたが、その効果は疑問視されている。 Inducing muscle hypertrophy by activating protein synthesis is expected as a treatment for muscle atrophy. It is very difficult to use general exercise therapy for patients with severe muscle atrophy and bedridden elderly people, and drug treatment is desired. So far, studies have been conducted to activate protein synthesis by administering Igf-1 (Non-patent Document 9) that activates the PI3K / Akt pathway, but its effect has been questioned.
本発明者の研究グループは、筋の不動化によってもたらされる筋萎縮では、神経性一酸化窒素シンターゼ(nNOS)が深く関与していることを明らかにしている(非特許文献3)。マウスの尾部懸垂時に筋細胞膜から遊離したnNOSは、細胞質に移って一酸化窒素(NO)を産生し、Forkhead box O(Foxo)のリン酸化を阻害してE3ユビキチンリガーゼの発現を活性化することにより筋萎縮を招いていた。またその後の研究により、nNOSは再負荷時における筋萎縮からの回復にも関与することがわかった。このことはnNOSが筋萎縮のみならず、筋肥大をも制御していることを示唆している。 The inventor's research group has clarified that neuronal nitric oxide synthase (nNOS) is deeply involved in muscle atrophy caused by muscle immobilization (Non-patent Document 3). NNOS released from the muscle cell membrane during mouse tail suspension moves to the cytoplasm, produces nitric oxide (NO), inhibits phosphorylation of Forkheadkbox O (Foxo), and activates expression of E3 ubiquitin ligase Caused muscle atrophy. Subsequent studies also showed that nNOS was involved in recovery from muscle wasting during reloading. This suggests that nNOS controls not only muscle atrophy but also muscle hypertrophy.
そこで本発明は、筋肥大時におけるnNOSの機能を明らかにするとともに、その分子基盤を元に新たな筋萎縮の治療法、すなわち筋肥大を促進するための方法及び手段を確立することを目的とする。 Therefore, the present invention aims to clarify the function of nNOS during muscle hypertrophy and to establish a new treatment method for muscle atrophy based on its molecular basis, that is, a method and means for promoting muscle hypertrophy. To do.
本発明者は、上記課題を解決するため鋭意検討を行った結果、NOとスーパーオキシドにより生成されるペルオキシ亜硝酸(peroxynitrite)がTRPチャネルを介して細胞内カルシウム濃度を制御することによって筋肥大が促進されることを見出し、スーパーオキシド及び/又はペルオキシ亜硝酸及び/又は筋細胞内のカルシウムの濃度を上昇させることによって筋肥大を促進することができるという知見を得、本発明を完成するに至った。 As a result of intensive studies to solve the above-mentioned problems, the present inventors have found that peroxynitrite produced by NO and superoxide controls intracellular calcium concentration via TRP channels, thereby causing muscle hypertrophy. It was found that it was promoted, and it was found that muscle hypertrophy can be promoted by increasing the concentration of superoxide and / or peroxynitrite and / or calcium in muscle cells, leading to the completion of the present invention. It was.
すなわち、本発明は、以下の[1]~[9]である。
[1]筋肥大を促進する物質又は因子のスクリーニング方法であって、
(a)被験物質又は因子による処置を受けた動物から得られた筋細胞において、スーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウムの濃度を測定するステップ、
(b)(a)の結果に基づいて被験物質又は因子を筋肥大を促進する物質又は因子の候補として同定するステップ
を含む方法。
[2]筋肥大を促進する物質又は因子のスクリーニング方法であって、
(a)筋細胞を、被験物質又は因子により処置するステップ、
(b)該筋細胞において、スーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウムの濃度を測定するステップ、
(c)(b)の結果に基づいて被験物質又は因子を筋肥大を促進する物質又は因子の候補として同定するステップ
を含む方法。
[3]被験物質又は因子による処置を行う前に、筋細胞において、スーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウム濃度を測定するステップをさらに含む、[1]又は[2]に記載の方法。
[4]筋細胞におけるスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウムの濃度が、非処置の筋細胞における濃度よりも高い場合には、被験物質又は因子が筋肥大を促進する物質又は因子の候補として同定される、[1]~[3]のいずれかに記載の方法。
[5]動物が、筋萎縮した動物、nNOS欠損型動物若しくは筋ジストロフィーを発症した動物である、又は筋細胞が、該動物に由来する筋細胞である、[1]~[4]のいずれかに記載の方法。
[6]スーパーオキシド供与剤、ペルオキシ亜硝酸供与剤及びTRPチャネルアゴニストからなる群より選択される少なくとも1種の薬物を含むことを特徴とする筋肥大促進剤。
[7]ペルオキシ亜硝酸供与剤が、SIN-1又はmolsidomineである、[6]に記載の筋肥大促進剤。
[8]TRPチャネルアゴニストがHyp9又はオルバニルである、[6]に記載の筋肥大促進剤。
[9]スーパーオキシド供与剤、ペルオキシ亜硝酸供与剤及びTRPチャネルアゴニストからなる群より選択される少なくとも1種の薬物の有効量を被験体に投与することを含む筋肥大促進方法。
That is, the present invention includes the following [1] to [9].
[1] A screening method for substances or factors that promote muscle hypertrophy,
(A) measuring the concentration of superoxide and / or peroxynitrite and / or calcium in muscle cells obtained from an animal treated with a test substance or factor;
(B) A method comprising identifying a test substance or factor as a candidate substance or factor that promotes muscle hypertrophy based on the result of (a).
[2] A screening method for substances or factors that promote muscle hypertrophy,
(A) treating muscle cells with a test substance or factor;
(B) measuring the concentration of superoxide and / or peroxynitrite and / or calcium in the muscle cells;
(C) A method comprising identifying a test substance or factor as a candidate substance or factor that promotes muscle hypertrophy based on the result of (b).
[3] The method according to [1] or [2], further comprising a step of measuring superoxide and / or peroxynitrite and / or calcium concentration in muscle cells before treatment with the test substance or factor. .
[4] When the concentration of superoxide and / or peroxynitrite and / or calcium in muscle cells is higher than that in untreated muscle cells, the test substance or factor is a substance or factor that promotes muscle hypertrophy. The method according to any one of [1] to [3], which is identified as a candidate.
[5] The animal according to any one of [1] to [4], wherein the animal is a muscle-atrophic animal, an nNOS-deficient animal, or an animal that has developed muscular dystrophy, or the muscle cell is a muscle cell derived from the animal. The method described.
[6] A muscle hypertrophy promoter comprising at least one drug selected from the group consisting of a superoxide donor, a peroxynitrite donor, and a TRP channel agonist.
[7] The muscle hypertrophy promoter according to [6], wherein the peroxynitrite donor is SIN-1 or molsidomine.
[8] The muscle hypertrophy promoter according to [6], wherein the TRP channel agonist is Hyp9 or olvanil.
[9] A method for promoting muscle hypertrophy, comprising administering to a subject an effective amount of at least one drug selected from the group consisting of a superoxide donor, a peroxynitrite donor, and a TRP channel agonist.
本発明に係るスクリーニング方法により、筋肥大を促進する物質又は因子を同定することができ、筋委縮などの治療法の開発に有用である。また、本発明に係る筋肥大促進剤は、筋肥大を効果的に促進することができ、筋委縮などの予防又は治療に有用である。 The screening method according to the present invention can identify substances or factors that promote muscle hypertrophy and is useful for the development of therapeutic methods such as muscle atrophy. The muscle hypertrophy promoter according to the present invention can effectively promote muscle hypertrophy and is useful for prevention or treatment of muscle atrophy.
以下、本発明を詳細に説明する。本願は、2011年9月14日に出願された日本国特許出願第2011-200716号の優先権を主張するものであり、上記特許出願の明細書及び/又は図面に記載される内容を包含する。 Hereinafter, the present invention will be described in detail. This application claims the priority of Japanese Patent Application No. 2011-200716 filed on September 14, 2011, and includes the contents described in the specification and / or drawings of the above patent application. .
本発明は、筋肥大におけるスーパーオキシド、ペルオキシ亜硝酸及び細胞内カルシウムの関与に基づいて、筋肥大を促進する物質及び因子をスクリーニングする方法と筋肥大促進剤を提供する。 The present invention provides a method and a muscle hypertrophy promoter for screening for substances and factors that promote muscle hypertrophy based on the involvement of superoxide, peroxynitrite and intracellular calcium in muscle hypertrophy.
本発明者は、筋肥大におけるnNOSの機能を明らかにするため、nNOS野生型及び欠損型マウスに後肢共働筋切除を行い、過負荷による代償性筋肥大を誘導した。同マウスの術後7日の筋重量を計測した結果、野生型は対照群と比べ40%程度筋重量が増加したが、欠損型は20%程度しか増加しなかった。nNOSが筋肥大過程のどの時期に機能するか明らかにするため、NOS活性測定を行った結果、nNOSは過負荷開始3分後に活性化し、1時間後には定常レベルにまで活性が低下した。nNOS活性を薬理学的に阻害した結果、この過負荷開始1時間の間に産生されたNOが筋肥大を促進することがわかった。nNOS野生型では過負荷3分後にmTORが活性化され、タンパク質合成経路が活性化していたが、欠損型ではその活性化が見られなかった(実施例1、及び伊藤ら, 第33回日本分子生物学会 演題2T9-11)。 In order to elucidate the function of nNOS in muscle hypertrophy, the present inventor performed hindlimb cooperative muscle excision in nNOS wild-type and deficient mice to induce compensatory muscle hypertrophy due to overload. As a result of measuring the muscle weight of the mice on the 7th day after the operation, the wild type increased the muscle weight by about 40% compared to the control group, but the deficient type increased only about 20%. In order to clarify when nNOS functions during muscle hypertrophy, NOS activity was measured. As a result, nNOS was activated 3 minutes after overloading and decreased to a steady level after 1 hour. As a result of pharmacological inhibition of nNOS activity, it was found that NO produced during 1 hour of overload promoted muscle hypertrophy. In the nNOS wild type, mTOR was activated 3 minutes after overload, and the protein synthesis pathway was activated, but in the deficient type, the activation was not observed (Example 1, and Ito et al., Sakai 33rd Japan Molecule). Biology Society Presentation 2T9-11).
しかし、血管拡張剤、8Br-cGMP等を投与しても欠損型の表現型を回復することはできず、nNOSによって産生されたNOは古典的なNO-cGMP経路によって制御される血管拡張を介さず、筋肥大を促進することが示唆された。そこでcGMP非依存的な経路であり、NOとスーパーオキシド(superoxide)との反応物であるペルオキシ亜硝酸(peroxynitrite)が筋肥大を促進する可能性があると考え、過負荷直後におけるsuperoxide、及びperoxynitriteの機能を明らかにする実験を行った(実施例2)。 However, administration of vasodilators such as 8Br-cGMP cannot restore the defective phenotype, and NO produced by nNOS is mediated by vasodilation controlled by the classical NO-cGMP pathway. It was suggested that muscle hypertrophy was promoted. Therefore, it is a cGMP-independent pathway, and it is thought that peroxynitrite, which is a reaction product of NO and superoxide, may promote muscle hypertrophy. Superoxide and peroxynitrite immediately after overload An experiment was conducted to clarify the function of (Example 2).
骨格筋におけるsuperoxideの主な供給分子であるNOXの筋肥大における機能を明らかにするため、NOX活性を薬理学的に阻害した。その結果、nNOS欠損型と同様に筋肥大の進行が抑えられた。NOとsuperoxideとの反応物であるperoxynitriteの筋肥大における機能を明らかにするため、superoxide消去剤又はperoxynitrite消去剤を投与した結果、nNOS欠損型と同様に筋肥大の進行が抑えられた。またperoxynitrite消去剤投与群におけるタンパク質合成経路の活性化を調べた結果、nNOS欠損型と同様、過負荷直後においてmTORの活性化が見られなかった。これらの結果から、nNOS由来のNOとNOX由来のsuperoxideから成るperoxynitriteが過負荷直後に産生され、mTORによって制御されるタンパク質合成経路を活性化させることで筋肥大を促進することを見出した。 In order to elucidate the function of NOX, the main supply molecule of superoxide in skeletal muscle, in muscle hypertrophy, NOX activity was pharmacologically inhibited. As a result, the progression of muscle hypertrophy was suppressed as in the case of the nNOS deficient type. In order to clarify the function of peroxynitrite, which is a reaction product of NO and superoxide, in hypertrophy, as a result of administering superoxide elimination agent or peroxynitrite elimination agent, the progression of muscle hypertrophy was suppressed as in the case of nNOS deficient type. Moreover, as a result of investigating the activation of the protein synthesis pathway in the peroxynitrite scavenger administration group, mTOR activation was not observed immediately after overloading, as in the case of the nNOS deficient type. From these results, it was found that peroxynitrite consisting of nNOS-derived NO and NOX-derived superoxide was produced immediately after overloading, and promoted muscle hypertrophy by activating the protein synthesis pathway controlled by mTOR.
さらに、NOX由来のsuperoxide、またperoxynitriteは細胞内カルシウム濃度を制御し得ることから、peroxynitriteが細胞内カルシウム濃度の制御を介してmTORを制御し、筋肥大を促進すると考えた(Adachi T et al. Nat Med 2004; 10: 1200-1207; Martins AS et al. J Physiol 2008; 286: 197-210)。細胞内カルシウム濃度とmTORの活性化の関係を明らかにするため、筋小胞体によるカルシウムの取り込みを阻害し、細胞内カルシウム濃度を上昇させるthapsigarginを筋注した結果、mTORの活性化が見られた。細胞内カルシウム濃度と筋肥大の関係を明らかにするため、カルシウムキレート剤であるBAPTA-AM、EGTAを筋注した結果、術後7日の筋重量が有意に抑制された。また骨格筋において代表的なカルシウムチャネルであるリアノジン受容体、DHPR、TRPチャネルの阻害薬(dantrolene, nifedipine, BTP2)を筋注した結果、TRPチャネル阻害薬BTP2によって術後7日の筋重量が有意に抑制されたが、dantrolene、nifedipineによっては抑制されなかった。BTP2は過負荷直後におけるmTORの活性化も抑制した。また、nNOS欠損型の表現型はNO供与剤、peroxynitrite供与剤によって回復されるが、その効果はBAPTA-AM、BTP2との共投与により完全に抑制された。さらに、TRPチャネルアゴニスト(Hyp9, olvanil)の投与により、nNOS欠損型の表現型は回復し、筋重量が増大した。またさらに、peroxynitriteが筋芽細胞におけるカルシウム濃度の上昇に関与しており、具体的には、筋小胞体のカルシウム放出を誘導することがわかった。 Furthermore, since superoxide derived from NOX and peroxynitrite can control the intracellular calcium concentration, we thought that peroxynitrite regulates mTOR through the control of intracellular calcium concentration and promotes muscle hypertrophy (Adachi T et al. Nat Med 2004; 10: 1200-1207; Martins AS et al. J Physiol 2008; 286: 197-210). In order to clarify the relationship between intracellular calcium concentration and mTOR activation, intramuscular injection of thapsigargin that inhibits calcium uptake by sarcoplasmic reticulum and raises intracellular calcium concentration resulted in activation of mTOR . In order to clarify the relationship between intracellular calcium concentration and muscle hypertrophy, BAPTA-AM and EGTA, calcium chelators, were injected intramuscularly. As a result, muscle weight was significantly suppressed on the 7th postoperative day. In addition, as a result of intramuscular injection of ryanodine receptor, DHPR, and TRP channel inhibitors (dantrolene, nifedipine, BTP2), which are typical calcium channels in skeletal muscle, the muscle weight on the 7th day after surgery was significant due to the TRP channel inhibitor BTP2. However, it was not suppressed by dantrolene or nifedipine. BTP2 also suppressed mTOR activation immediately after overload. Moreover, the nNOS-deficient phenotype was restored by NO donor and peroxynitrite donor, but the effect was completely suppressed by co-administration with BAPTA-AM and BTP2. Furthermore, administration of a TRP channel agonist (Hyp9, olvanil) restored the nNOS-deficient phenotype and increased muscle weight. Furthermore, it was found that peroxynitrite is involved in the increase of calcium concentration in myoblasts, specifically, inducing calcium release in the sarcoplasmic reticulum.
これらの結果から、superoxide、peroxynitrite及びその下流の細胞内カルシウム濃度制御が、過負荷によるタンパク質合成経路の活性化に重要であることが明らかになった。またperoxynitriteはTRPチャネルによる細胞内カルシウム濃度(特に筋小胞体内カルシウム濃度)の制御を介して、筋肥大を促進することが見出された。そのため、TRPチャネルのアゴニストを用い、細胞内カルシウム濃度を上昇させることで人為的にタンパク質合成を活性化させることが可能であると考えられる。 From these results, it became clear that control of superoxide, peroxynitrite and its downstream intracellular calcium concentration is important for the activation of protein synthesis pathway by overload. Peroxynitrite was also found to promote muscle hypertrophy through the regulation of intracellular calcium concentration (especially sarcoplasmic reticulum calcium concentration) by TRP channels. Therefore, it is considered possible to artificially activate protein synthesis by increasing the intracellular calcium concentration using an agonist of TRP channel.
本発明では、筋細胞におけるスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウムの濃度に基づいて、筋肥大を促進又は阻害する物質又は因子を同定する。すなわち、動物又は筋細胞を、被験物質又は因子で処置し、該動物又は筋細胞におけるスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウムの濃度を測定することによって、該被験物質又は因子がスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウムの濃度、すなわち筋肥大に影響を及ぼすか否かを判定することができる。 In the present invention, a substance or factor that promotes or inhibits muscle hypertrophy is identified based on the concentration of superoxide and / or peroxynitrite and / or calcium in muscle cells. That is, by treating an animal or muscle cell with a test substance or factor and measuring the concentration of superoxide and / or peroxynitrite and / or calcium in the animal or muscle cell, the test substance or factor becomes superoxide. And / or whether it affects peroxynitrite and / or calcium concentrations, ie muscle hypertrophy.
本発明に関連して、「筋肥大」とは、内在タンパク質量の増加に伴う単一筋線維重量又は断面積の増加による筋重量の増加といい、「筋肥大を促進する」とは、内在タンパク質量の増加を促進することによって、単一筋線維重量又は断面積の増加による筋重量の増加を促進することをいう。 In the context of the present invention, “muscular hypertrophy” refers to an increase in muscle weight due to an increase in the weight of a single muscle fiber or cross-sectional area accompanying an increase in the amount of endogenous protein, and “promotes muscle hypertrophy” By promoting the increase in amount, it means promoting the increase in muscle weight due to the increase in single muscle fiber weight or cross-sectional area.
本発明に係るスクリーニング方法(以下、「本スクリーニング方法」ともいう)においては、被験物質又は因子で処置された動物から得られた筋細胞を準備するか、あるいは筋細胞を被験物質又は因子で処置し、該筋細胞におけるスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウム濃度を測定する。好ましくは、被験物質又は因子による処置を行う前に、動物から得られた筋細胞又は筋細胞におけるスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウム濃度を測定する。 In the screening method according to the present invention (hereinafter, also referred to as “the screening method”), muscle cells obtained from an animal treated with a test substance or factor are prepared, or muscle cells are treated with the test substance or factor. Then, the superoxide and / or peroxynitrite and / or calcium concentration in the muscle cells is measured. Preferably, prior to treatment with a test substance or factor, superoxide and / or peroxynitrite and / or calcium concentrations in muscle cells or muscle cells obtained from animals are measured.
対象となる動物は、筋肉を有する動物であれば特に限定されるものではなく、哺乳動物、例えばヒト、霊長類(サル、チンパンジーなど)、家畜動物(ウシ、ウマ、ブタなど)、愛玩動物(イヌ、ネコなど)、実験動物(マウス、ラット、サルなど)や、両生類、爬虫類及び鳥類も含まれる。また動物は、正常な動物であってもよいし、あるいは筋萎縮した動物、nNOS欠損型動物又は筋ジストロフィーを発症した動物などであってもよい。一般的には、細胞レベルにおいて被験物質又は因子の有効性が確認された後に、実験動物、さらにはヒトにおいて、例えば臨床試験などにより有効性の評価が行われる。 The target animal is not particularly limited as long as it has muscles, and mammals such as humans, primates (monkeys, chimpanzees, etc.), livestock animals (cattle, horses, pigs, etc.), pets ( Dogs, cats, etc.), laboratory animals (mouse, rats, monkeys, etc.), amphibians, reptiles and birds. Further, the animal may be a normal animal, an muscularly atrophic animal, an nNOS-deficient animal, or an animal that has developed muscular dystrophy. In general, after the effectiveness of a test substance or factor is confirmed at the cellular level, the effectiveness is evaluated in a laboratory animal, and further in a human, for example, by a clinical test.
本スクリーニング方法の対象となる被験物質又は因子の種類は特に限定されるものではない。例えば、被験物質又は因子は、任意の物質、具体的には、天然に生じる分子、例えば、アミノ酸、ペプチド、オリゴペプチド、ポリペプチド、タンパク質、核酸、脂質、炭水化物(糖等)、ステロイド、グリコペプチド、糖タンパク質、プロテオグリカンなど;天然に生じる分子の合成アナログ又は誘導体、例えば、ペプチド擬態物、核酸分子(アプタマー、アンチセンス核酸、二本鎖RNA(RNAi)等)など;天然に生じない分子、例えば、コンビナトリアルケミストリー技術等を用いて作製した低分子有機化合物(無機及び有機化合物ライブラリー、又はコンビナトリアルライブラリー等)など;並びにそれらの混合物を挙げることができる。また被験物質又は因子は、単一物質であってもよいし、複数の物質から構成される複合体や、転写因子等であってもよい。さらに、被験物質又は因子は、放射線、紫外線、炭素濃度、温度などの環境因子であってもよい。 The type of test substance or factor that is the subject of this screening method is not particularly limited. For example, the test substance or factor can be any substance, specifically a naturally occurring molecule such as an amino acid, peptide, oligopeptide, polypeptide, protein, nucleic acid, lipid, carbohydrate (such as sugar), steroid, glycopeptide Synthetic analogs or derivatives of naturally occurring molecules, such as peptidomimetics, nucleic acid molecules (aptamers, antisense nucleic acids, double-stranded RNA (RNAi), etc.), etc .; non-naturally occurring molecules, such as And low molecular organic compounds (inorganic and organic compound libraries, combinatorial libraries, etc.) produced using combinatorial chemistry techniques and the like; and mixtures thereof. The test substance or factor may be a single substance, a complex composed of a plurality of substances, a transcription factor, or the like. Furthermore, the test substance or factor may be an environmental factor such as radiation, ultraviolet rays, carbon concentration, or temperature.
また、被験物質又は因子としては単一の被験物質又は因子を独立に試験しても、いくつかの候補となる被験物質又は因子の混合物(ライブラリーなどを含む)について試験をしてもよい。複数の被験物質又は因子を含むライブラリーとしては、合成化合物ライブラリー(コンビナトリアルライブラリーなど)、ペプチドライブラリー(コンビナトリアルライブラリーなど)などが挙げられる。 In addition, as a test substance or factor, a single test substance or factor may be tested independently, or a mixture of several candidate test substances or factors (including a library or the like) may be tested. Examples of the library containing a plurality of test substances or factors include a synthetic compound library (such as a combinatorial library) and a peptide library (such as a combinatorial library).
動物を被験物質又は因子で処置する場合には、その処置量、処置期間、処置経路などの処置条件は、被験物質又は因子の種類などにより異なるが、当業者であれば容易に決定することができる。例えば、動物に被験物質を投与する場合の投与経路は、被験物質の種類、使用する動物の種類などに応じて、筋肉内注射、経口投与、静脈注射、腹腔内注射、経皮投与、皮下注射等の投与形態を適宜使用することができる。 When an animal is treated with a test substance or factor, the treatment conditions such as the treatment amount, treatment period, treatment route, etc. vary depending on the type of test substance or factor, but those skilled in the art can easily determine it. it can. For example, when administering a test substance to an animal, the route of administration is intramuscular injection, oral administration, intravenous injection, intraperitoneal injection, transdermal administration, subcutaneous injection, depending on the type of test substance and the type of animal used. Such administration forms can be appropriately used.
筋細胞は、当技術分野で公知の方法により採取することができる。具体的には、例えば、骨格筋を細切し、50 ml円錐底チューブに移した後、筋重量1 gあたり4 mlのDispase2 (2.4 IU/ml) -Collagenase XI (0.2%) 溶液を加え、37℃、45~60分間インキュベートし、途中15分ごとにピペットを用いてピペッティングする。その後、18 Gの注射針に数回通して組織片を粉砕し上清を回収する。上清を80μmのフィルターに通して細胞塊を除き、細胞懸濁液を50 ml円錐底チューブに移し、さらにDispase2 (2.4 IU/ml) -Collagenase XI (0.2%) 処理後のチューブに増殖培地を加え、その上清を同様にフィルターに通して回収する。増殖培地を加え、合計30 mlとし、数回ピペッティングを行い、1,000 rpmで5分間、4℃で遠心する。上清を捨て沈殿細胞に増殖培地20 mlを加え、再度懸濁した後、1,000 rpmで5分間、4℃で遠心する。沈殿細胞を増殖培地25 mlに懸濁し、コーティングなしの15 cm培養皿に移しbFGFを添加し、37℃、5%CO2濃度で90分間培養後、未付着細胞を含む上清を回収する。この上清を用いて培養皿の底面を洗い、180度向きを変えてもう一度37℃、5%CO2濃度で90分間培養し、上清を回収する。回収した上清を15 cmコラーゲンコート培養皿に移しbFGFを添加し、37℃、5%CO2濃度で一晩培養する。翌日筋芽細胞が30~40%コンフルエント以上になっているようなら継代を行い、それ以下なら培地交換に留める。筋分化を防ぐために以後、毎日継代ないしは培地交換を行う。あるいは、市販の筋細胞又は公に入手可能な筋細胞を使用することも可能である。 Muscle cells can be collected by methods known in the art. Specifically, for example, after chopping skeletal muscle and transferring it to a 50 ml conical bottom tube, 4 ml of Dispase2 (2.4 IU / ml) -Collagenase XI (0.2%) solution per 1 g of muscle weight is added, Incubate at 37 ° C for 45-60 minutes, pipetting with a pipette every 15 minutes. Thereafter, the tissue piece is crushed by passing several times through an 18 G injection needle, and the supernatant is collected. Pass the supernatant through an 80 μm filter to remove cell clumps, transfer the cell suspension to a 50 ml conical bottom tube, and add growth medium to the tube treated with Dispase2 (2.4 IU / ml) -Collagenase XI (0.2%). In addition, the supernatant is similarly collected through a filter. Add growth medium to a total volume of 30 ml, pipette several times, and centrifuge at 1,000 rpm for 5 minutes at 4 ° C. Discard the supernatant, add 20 ml of growth medium to the precipitated cells, resuspend, and centrifuge at 1,000 rpm for 5 minutes at 4 ° C. The precipitated cells are suspended in 25 ml of growth medium, transferred to an uncoated 15 cm culture dish, added with bFGF, and cultured for 90 minutes at 37 ° C., 5% CO 2 concentration, and the supernatant containing unattached cells is collected. Using this supernatant, wash the bottom of the culture dish, change the direction 180 degrees, incubate again at 37 ° C, 5% CO 2 concentration for 90 minutes, and collect the supernatant. Transfer the collected supernatant to a 15 cm collagen-coated culture dish, add bFGF, and culture overnight at 37 ° C. and 5% CO 2 concentration. The next day, if the myoblasts are more than 30-40% confluent, passaging, and if less, change the medium. In order to prevent myogenic differentiation, subculture or medium exchange is performed every day thereafter. Alternatively, commercially available muscle cells or publicly available muscle cells can be used.
筋細胞を被験物質又は因子と接触させる場合、その接触の条件は、その物質又は因子の種類により異なるが、当業者であれば容易に決定することができる。例えば、そのような接触は、筋細胞を被験物質を添加した培地中で培養することにより、筋細胞を被験物質を含む溶液中に浸漬することにより、筋細胞上に被験物質を積層することにより、又は筋細胞を被験因子の存在下で培養することにより行うことができる。 When the muscle cell is brought into contact with the test substance or factor, the contact condition varies depending on the type of the substance or factor, but can be easily determined by those skilled in the art. For example, such contact can be achieved by culturing myocytes in a medium supplemented with a test substance, immersing the myocytes in a solution containing the test substance, and laminating the test substance on the muscle cells. Alternatively, it can be performed by culturing myocytes in the presence of a test factor.
また、被験物質又は因子の効果及び有効性は、いくつかの条件で検討することも可能である。そのような条件としては、被験物質又は因子で処置する時間又は期間、量(大小)、回数などが挙げられる。例えば、被験物質の希釈系列を調製するなどして複数の用量を設定することができる。被験物質又は因子の処置期間も適宜設定することができるが、例えば、1日から数週間、数ヶ月、数年の期間にわたって処置を行うことができる。 Also, the effect and effectiveness of the test substance or factor can be examined under several conditions. Such conditions include time or duration, amount (large or small), number of times, etc. of treatment with the test substance or factor. For example, a plurality of doses can be set by preparing a dilution series of the test substance. The treatment period of the test substance or factor can also be set as appropriate. For example, the treatment can be performed over a period of 1 day to several weeks, months, and years.
さらに、複数の物質及び/又は因子の相加作用、相乗作用などを検討する場合には、被験物質及び/又は因子を組み合わせて用いてもよい。 Furthermore, when examining the additive action, synergistic action, etc. of a plurality of substances and / or factors, the test substances and / or factors may be used in combination.
続いて、動物又は筋細胞を被験物質又は因子で処置した後、適当な時期に、該動物から得られた筋細胞又は該筋細胞におけるスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウムの濃度を測定する。例えば、処置の直後、30分後、1時間後、3時間後、5時間後、10時間後、15時間後、20時間後、24時間(1日)後、2~10日後、10~20日後、20~30日後、1ヶ月~6ヵ月後に測定を行う。 Subsequently, after the animal or muscle cell is treated with the test substance or factor, the concentration of superoxide and / or peroxynitrite and / or calcium in the muscle cell obtained from the animal or the muscle cell is appropriately determined. taking measurement. For example, immediately after treatment, 30 minutes, 1 hour, 3 hours, 5 hours, 10 hours, 15 hours, 20 hours, 24 hours (1 day), 2-10 days, 10-20 Measurements are taken after 20-30 days, 1-6 months later.
スーパーオキシドの測定は、当技術分野で公知の方法により行うことができ、例えばスーパーオキシドと反応することにより発光するルシゲニンなどを使用して、その発光強度を測定することにより行うことができる。 The measurement of superoxide can be performed by a method known in the art, for example, by using lucigenin that emits light by reacting with superoxide, and measuring the emission intensity.
ペルオキシ亜硝酸の測定は、当技術分野で公知の方法により行うことができ、例えばペルオキシ亜硝酸が活性酸素種又は活性窒素種と反応することにより発せられる蛍光物質の蛍光強度を測定し、ペルオキシ亜硝酸消去剤処理した対照群との差を算出することにより行うことができる。 Peroxynitrite can be measured by a method known in the art. For example, the peroxynitrite is measured by measuring the fluorescence intensity of a fluorescent substance emitted by reacting peroxynitrite with an active oxygen species or an active nitrogen species. This can be done by calculating the difference from the control group treated with the nitric acid scavenger.
また筋細胞内のカルシウム濃度の測定も当技術分野で公知の方法により行うことができ、例えば、カルシウムと結合することにより蛍光を発するFluo-4などを使用して、その蛍光強度を測定することにより行うことができる。 In addition, the calcium concentration in muscle cells can also be measured by a method known in the art. For example, the fluorescence intensity is measured using Fluo-4 that emits fluorescence when bound to calcium. Can be performed.
筋細胞におけるスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウム濃度を測定した後、対照と比較し、スーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウム濃度を上昇又は低減させる被験物質又は因子を、筋肥大を促進又は阻害する物質又は因子として選択する。対照としては、被験物質又は因子による処置を受けていない動物又は筋細胞などを用いることができる。 A test substance or factor that increases or decreases the superoxide and / or peroxynitrite and / or calcium concentration relative to the control after measuring the superoxide and / or peroxynitrite and / or calcium concentration in muscle cells, Selected as a substance or factor that promotes or inhibits muscle hypertrophy. As a control, animals or muscle cells that have not been treated with the test substance or factor can be used.
また、本発明においては、一次スクリーニングとして、筋細胞を被験物質又は因子で処置し、該筋細胞におけるスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウムの濃度の上昇を示した被験物質又は因子を選択し、次に二次スクリーニングとして、選択した被験物質又は因子で動物を処置し、該動物の筋細胞におけるスーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウム濃度を測定して濃度上昇を示した被験物質又は因子を選択してもよい。 Moreover, in the present invention, as a primary screening, a test substance or factor that has been treated with a test substance or factor and that has increased the superoxide and / or peroxynitrite and / or calcium concentration in the myocyte is obtained. Selected and then as a secondary screen, animals were treated with the selected test substance or factor and the concentrations of superoxide and / or peroxynitrite and / or calcium in the muscle cells of the animals were measured to show increased concentrations A test substance or factor may be selected.
さらに、筋肥大を促進する物質又は因子のスクリーニングにおいては、選択された被験物質又は因子を、実験動物に投与して、被験物質又は因子が実験動物における筋肥大を促進するか否かを判定してもよい。実験動物としては、後肢懸垂、除神経、デキサメタゾン投与等により筋萎縮が誘導されたモデル動物、好ましくはマウスを用いることができる。被験物質又は因子が実験動物における筋肥大を促進するか否かの判定は、実験動物の種類などにより異なるが、当業者であれば、当技術分野で公知の方法により適宜判定することができる。例えば、動物から筋組織を採取し、その筋重量を測定することができる。 Further, in screening for a substance or factor that promotes muscle hypertrophy, the selected test substance or factor is administered to an experimental animal to determine whether the test substance or factor promotes muscle hypertrophy in the experimental animal. May be. As the experimental animal, a model animal, preferably a mouse, in which muscle atrophy has been induced by hindlimb suspension, denervation, dexamethasone administration, or the like can be used. Whether or not a test substance or factor promotes muscle hypertrophy in an experimental animal depends on the type of experimental animal and the like, but can be appropriately determined by those skilled in the art by methods known in the art. For example, muscle tissue can be collected from an animal and the muscle weight can be measured.
以上のようにして、本スクリーニング方法により、筋肥大を促進する物質又は因子の候補を同定し、さらには筋肥大を促進する物質又は因子の有効性を確認することができる。 As described above, by this screening method, a candidate substance or factor that promotes muscle hypertrophy can be identified, and further, the effectiveness of the substance or factor that promotes muscle hypertrophy can be confirmed.
本発明に係る筋肥大促進剤は、スーパーオキシド及び/又はペルオキシ亜硝酸及び/又はカルシウム濃度を上昇させることができる物質又は因子を含むものである。具体的には、本発明に係る筋肥大促進剤は、スーパーオキシド供与剤、ペルオキシ亜硝酸供与剤及びTRPチャネルアゴニストからなる群より選択される少なくとも1種の薬物を含む。本発明に係る筋肥大促進剤は、筋萎縮などの疾患又は障害の予防又は治療、特に重度の筋萎縮を伴い、リハビリテーション等の運動療法が困難な患者の治療や寝たきりの患者の筋増量に有用である。 The muscle hypertrophy promoter according to the present invention contains a substance or factor capable of increasing the superoxide and / or peroxynitrite and / or calcium concentration. Specifically, the muscle hypertrophy promoter according to the present invention includes at least one drug selected from the group consisting of a superoxide donor, a peroxynitrite donor, and a TRP channel agonist. The muscle hypertrophy promoter according to the present invention is useful for the prevention or treatment of diseases or disorders such as muscle atrophy, especially for the treatment of patients with severe muscle atrophy and difficulty in exercise therapy such as rehabilitation and for the increase in muscle mass of bedridden patients. It is.
有効成分であるスーパーオキシド供与剤としては、当技術分野においてスーパーオキシドを供与することが知られている薬物であれば任意の薬物を用いることができる。 As the superoxide donor which is an active ingredient, any drug can be used as long as it is a drug known to donate superoxide in this technical field.
有効成分であるペルオキシ亜硝酸供与剤としては、当技術分野においてペルオキシ亜硝酸を供与することが知られている薬物であれば任意の薬物を用いることができる。例えば、SIN-1、モルシドミン(molsidomine)などが含まれ、いずれも市販されている。また有効成分であるTRPチャネルアゴニストも、当技術分野においてTRPチャネルに対してアゴニスト活性を有することが知られている薬物であれば任意の薬物を用いることができる。例えば、Hyp9、オルバニルなどが含まれ、市販品を用いることができる。 As the peroxynitrite donating agent that is an active ingredient, any drug can be used as long as it is a drug known to donate peroxynitrite in this technical field. For example, SIN-1, molsidomine, etc. are included and all are commercially available. As the active ingredient TRP channel agonist, any drug can be used as long as it is a drug known in the art to have agonist activity with respect to the TRP channel. For example, Hyp9, olvanil and the like are included, and commercially available products can be used.
本筋肥大促進剤は、有効成分として、1種のスーパーオキシド供与剤及び/又はペルオキシ亜硝酸供与剤及び/又はTRPチャネルアゴニストを含有するものであってもよいし、又は2種以上のスーパーオキシド供与剤及び/又はペルオキシ亜硝酸供与剤及び/又はTRPチャネルアゴニストを組み合わせて含有するものであってもよい。本筋肥大促進剤は、スーパーオキシド供与剤、ペルオキシ亜硝酸供与剤及びTRPチャネルアゴニストからなる群より選択される少なくとも1種の薬物を有効成分として含有する限り、上述した筋肥大の促進作用を発揮する。 The muscle hypertrophy promoter may contain, as an active ingredient, one superoxide donor and / or a peroxynitrite donor and / or a TRP channel agonist, or two or more superoxide donors And / or a combination of a peroxynitrite donating agent and / or a TRP channel agonist. As long as this muscle hypertrophy promoter contains as an active ingredient at least one drug selected from the group consisting of a superoxide donor, a peroxynitrite donor, and a TRP channel agonist, it exerts the above-described muscle hypertrophy promoter action. .
本筋肥大促進剤は、有効成分であるスーパーオキシド供与剤及び/又はペルオキシ亜硝酸供与剤及び/又はTRPチャネルアゴニストの他、薬学的に許容される担体又は添加物を共に含むものであってもよい。このような担体及び添加物の例として、水、薬学的に許容される有機溶剤、コラーゲン、ポリビニルアルコール、ポリビニルピロリドン、カルボキシビニルポリマー、アルギン酸ナトリウム、水溶性デキストラン、カルボキシメチルスターチナトリウム、ペクチン、キサンタンガム、アラビアゴム、カゼイン、ゼラチン、寒天、グリセリン、プロピレングリコール、ポリエチレングリコール、ワセリン、パラフィン、ステアリルアルコール、ステアリン酸、ヒト血清アルブミン、マンニトール、ソルビトール、ラクトースなどが挙げられる。使用される添加物は、剤形に応じて上記の中から適宜又は組み合わせて選択される。 The muscle hypertrophy promoter may contain a pharmaceutically acceptable carrier or additive in addition to a superoxide donor and / or a peroxynitrite donor and / or a TRP channel agonist as active ingredients. . Examples of such carriers and additives include water, pharmaceutically acceptable organic solvents, collagen, polyvinyl alcohol, polyvinyl pyrrolidone, carboxyvinyl polymer, sodium alginate, water-soluble dextran, sodium carboxymethyl starch, pectin, xanthan gum, Examples include gum arabic, casein, gelatin, agar, glycerin, propylene glycol, polyethylene glycol, petrolatum, paraffin, stearyl alcohol, stearic acid, human serum albumin, mannitol, sorbitol, and lactose. The additive to be used is appropriately or in combination selected from the above depending on the dosage form.
本筋肥大促進剤を経口投与する場合は、錠剤、カプセル剤(硬カプセル剤、軟カプセル剤、マイクロカプセルなど)、顆粒剤、散剤、丸剤、トローチ剤、内用水剤、液剤、懸濁剤、乳剤、シロップ剤などのいずれのものであってもよく、使用する際に再溶解させる乾燥製剤にしてもよい。また、本筋肥大促進剤を非経口投与する場合は、例えば静脈内注射(点滴を含む)、筋肉内注射、腹腔内注射及び皮下注射用の注射剤(例えば溶液、乳剤、懸濁剤)、軟膏剤、クリーム剤、座剤、パップ剤、吸入剤、リニメント剤、エアゾル剤などの外用剤などの製剤形態を選択することができ、注射剤の場合は単位用量アンプル又は多用量容器の状態で提供される。 When this muscle hypertrophy promoter is administered orally, tablets, capsules (hard capsules, soft capsules, microcapsules, etc.), granules, powders, pills, troches, liquids for internal use, liquids, suspensions, Any of emulsion, syrup, etc. may be used, and it may be a dry preparation re-dissolved when used. In addition, when the muscle hypertrophy promoter is administered parenterally, for example, intravenous injection (including infusion), intramuscular injection, intraperitoneal injection and subcutaneous injection (eg, solution, emulsion, suspension), ointment Preparations such as pills, creams, suppositories, cataplasms, inhalants, liniments, aerosols and other external preparations can be selected. In the case of injections, they are provided in unit dose ampoules or multi-dose containers Is done.
これらの各種製剤は、医薬において通常用いられる賦形剤、増量剤、結合剤、湿潤剤、崩壊剤、滑沢剤、界面活性剤、分散剤、緩衝剤、pH調整剤、保存剤、溶解補助剤、防腐剤、矯味矯臭剤、吸収促進剤、無痛化剤、安定化剤、等張化剤などを適宜選択し、常法により製造することができる。 These various preparations are excipients, extenders, binders, wetting agents, disintegrating agents, lubricants, surfactants, dispersants, buffering agents, pH adjusting agents, preservatives, solubilizers commonly used in medicine. Agents, preservatives, flavoring agents, absorption promoters, soothing agents, stabilizers, tonicity agents and the like can be appropriately selected and produced by conventional methods.
本筋肥大促進剤に配合するスーパーオキシド供与剤及び/又はペルオキシ亜硝酸供与剤及び/又はTRPチャネルアゴニストは、その有効成分の種類、用途、剤形、投与経路などにより異なるが、例えば総重量を基準として0.01~90重量%、好ましくは1~50重量%である。 The superoxide donor and / or peroxynitrite donor and / or TRP channel agonist to be added to the muscle hypertrophy promoter varies depending on the type of active ingredient, application, dosage form, route of administration, etc. 0.01 to 90% by weight, preferably 1 to 50% by weight.
また、本筋肥大促進剤の有効量(投与量又は摂取量)は、該剤に含まれる有効成分の種類、被験体の年齢及び体重、投与経路、投与回数により異なり、広範囲に変更することができる。例えば、通常成人1日当たりのスーパーオキシド供与剤及び/又はペルオキシ亜硝酸供与剤及び/又はTRPチャネルアゴニストの有効量を、1日1回又は数回に分けて、約1週間~約1年間、好ましくは約1ヶ月~約12ヶ月にわたり投与することができる。 In addition, the effective amount (dose or intake) of the muscle hypertrophy promoter varies depending on the type of active ingredient contained in the agent, the age and weight of the subject, the administration route, and the number of administrations, and can be varied over a wide range. . For example, the effective amount of a superoxide donor and / or peroxynitrite donor and / or TRP channel agonist per day for an adult is usually about 1 week to about 1 year, preferably once or several times a day. Can be administered for about 1 month to about 12 months.
本筋肥大促進剤は、使用する対象(被験体)を特に限定するものではない。例えば、ヒト、家畜(ウシなど)、愛玩動物(イヌ、ネコなど)、実験動物(サルなど)などの被験体に投与する又は摂取させることができる。 This muscle hypertrophy promoter does not particularly limit the target (subject) to be used. For example, it can be administered to or ingested by subjects such as humans, domestic animals (such as cows), pets (such as dogs and cats), and laboratory animals (such as monkeys).
本筋肥大促進剤は、当技術分野で公知の他の筋肥大促進剤又は筋肥大促進に有効な方法と組み合わせることができる。例えば、筋肥大に必要な栄養素であるタンパク質の投与、該栄養素を筋に同化させるホルモン(成長ホルモンなど)の投与、筋肉への負荷(例えば軽度な運動、筋力トレーニング、加圧トレーニング)などと組み合わせることができる。 This muscle hypertrophy promoter can be combined with other muscle hypertrophy promoters known in the art or methods effective for promoting muscle hypertrophy. For example, it is combined with administration of protein that is a nutrient necessary for muscle hypertrophy, administration of hormones (growth hormone, etc.) that assimilate the nutrient to the muscle, load on the muscle (eg, mild exercise, strength training, pressure training), etc. be able to.
本筋肥大促進剤は、医薬組成物としての用途に限定されず、その他、例えば食品又は飼料などに配合されてもよい。「食品」及び「飼料」とは、栄養素を1種以上含む天然物及びその加工品をいい、あらゆる飲食物を含む。本筋肥大促進剤が配合された食品又は飼料は、筋肥大を促進するための健康補助製品として有用である。 The present muscle hypertrophy promoter is not limited to the use as a pharmaceutical composition, and may be blended in other products such as food or feed. “Food” and “feed” refer to natural products containing one or more nutrients and processed products thereof, including all food and drink. A food or feed containing the present muscle hypertrophy promoter is useful as a health supplement product for promoting muscle hypertrophy.
本筋肥大促進剤を食品に配合する場合、固体状食品、ゼリー状食品、液状食品、カプセル状食品など様々な形態の食品に添加することができる。ここで、固体状食品としては、パン生地;せんべい、ビスケット、クッキーなどの焼き菓子用生地;そば、うどんなどの麺類;かまぼこ、ちくわなどの魚肉製品;ハム、ソーセージなどの畜肉製品;粉ミルクなどが挙げられる。また、ゼリー状食品としては、フルーツゼリー;コーヒーゼリーなどが挙げられる。さらに、液状食品としては、清涼飲料・果実飲料などの飲料類(茶、コーヒー、紅茶、発酵乳、乳酸菌飲料など)、調味料など(マヨネーズ、ドレッシング、味付け調味液など)が挙げられる。カプセル状食品としては、ハードカプセル、ソフトカプセルなどがあげられる。 When adding this muscle hypertrophy promoter to food, it can be added to various forms of food such as solid food, jelly food, liquid food, and capsule food. Here, solid food includes bread dough; dough for baked confectionery such as rice crackers, biscuits and cookies; noodles such as buckwheat and udon; fish products such as kamaboko and chikuwa; livestock products such as ham and sausage; powdered milk and the like It is done. Examples of the jelly-like food include fruit jelly and coffee jelly. Furthermore, examples of liquid foods include beverages such as soft drinks and fruit beverages (tea, coffee, tea, fermented milk, lactic acid bacteria beverages, etc.), seasonings (mayonnaise, dressings, seasoning seasonings, etc.). Examples of capsule foods include hard capsules and soft capsules.
本筋肥大促進剤を食品に添加する場合、添加量としては、食品全体に対してスーパーオキシド供与剤及び/又はペルオキシ亜硝酸供与剤及び/又はTRPチャネルアゴニストの含有量が0.01~90重量%となるように配合することができる。効果が期待できる摂取量は年齢、体重、性別、症状の程度などを考慮して、個々の場合に応じて適宜決定される。摂取回数は一日に何回かに分けて摂取できるが、その場合は、回数に応じて量を分割することも可能である。また、長期にわたり連続して摂取することが可能である。 When the muscle hypertrophy promoter is added to food, the amount added is 0.01 to 90% by weight of the superoxide donor and / or peroxynitrite donor and / or TRP channel agonist with respect to the whole food. It can mix | blend as follows. The intake that can be expected to be effective is appropriately determined according to the individual case, taking into account the age, weight, sex, and degree of symptoms. The number of intakes can be divided into several times a day, in which case the amount can be divided according to the number of times. In addition, it can be taken continuously over a long period of time.
以下、本発明を実施例及び図面によりさらに具体的に説明する。ただし、以下の実施例は、本発明を限定するものではない。 Hereinafter, the present invention will be described more specifically with reference to examples and drawings. However, the following examples do not limit the present invention.
[実施例1]
本実施例では、筋肥大におけるnNOSの寄与を明らかにするため、nNOS野生型及び欠損型マウスに共働筋切除を行い、過負荷による代償性筋肥大を誘導した。
対象には、Jackson Laboratories(Wilmington, MA)より購入したnNOS欠損型マウス、及び日本クレア(日本)より購入したC57BL/6マウスを用いた。
[Example 1]
In this example, in order to clarify the contribution of nNOS in muscle hypertrophy, cooperative muscle excision was performed on nNOS wild-type and deficient mice to induce compensatory muscle hypertrophy due to overload.
Subjects include Jackson Laboratories (Wilmington, NNOS-deficient mice purchased from MA) and C57BL / 6 mice purchased from CLEA Japan (Japan) were used.
以下のように共働筋切除による代償性筋肥大誘導と薬物投与を行った。すなわち、12週齢マウスの腓腹筋及びヒラメ筋の腱を麻酔下で切除した(Adams, G.R. and F. Haddad, J Appl Physiol, 1996. 81(6):2509-16)。対照群には皮膚を切る偽手術(sham)を施した。術後は飲料水に1 mg/mlアンピシリンを加えた。NOS阻害剤L-NAME(1mg/ml, Sigma-Aldrich)は飲水投与し、飲水は毎日交換した。nNOS阻害剤7-NI(50 mg/kg, Calbiochem)及びNOX阻害剤DPI(2 mg/kg, Sigma-Aldrich)は過負荷開始30分前若しくは60分後に腹腔内投与した。また、apocynin(10 mg/kg, Sigma-Aldrich)、tadalafil(10 mg/kg, Toronto Research Chemicals)、sildenafil(10 mg/kg, Sigma-Aldrich)、8-Br-cGMP(10 mg/kg, Calbiochem)、rotenone(5 mg/kg, Sigma-Aldrich)、allopurinol(10 mg/kg, Cayman)、尿酸(200 mg/kg Sigma-Aldrich;実施例2)、FeTPPS(30 mg/kg, Calbiochem;実施例2)、ebselen(30 mg/kg, Sigma-Aldrich;実施例2)、molsidomine(50 mg/kg, Sigma-Aldrich;実施例2)、SIN-1(10 mg/kg, DOJINDO;実施例2及び3)、SNAP(20 mg/kg, Calbiochem;実施例3)は過負荷開始30分前に腹腔内投与した。BAPTA-AM(50μM, Calbiochem;実施例3)、EGTA(1 mM, Dojindo;実施例3)、dantrolene(10μM, Sigma-Aldrich;実施例3)、nifedipine(10μM, Sigma-Aldrich;実施例3)、BTP2(10μM, Calbiochem;実施例3)、Gd3+(50μm, Sigma-Aldrich;実施例3)、GsMTx-4(10μM, Wako;実施例3)、thapsigargin(20μM, Calbiochem;実施例3)、Hyp9(10μM, Sigma-Aldrich;実施例3)、olvanil(10μM, Sigma-Aldrich;実施例3)は過負荷開始30分前に筋注した。 Compensatory muscle hypertrophy induction and drug administration by synergistic muscle resection were performed as follows. That is, the gastrocnemius and soleus tendons of 12-week-old mice were excised under anesthesia (Adams, GR and F. Haddad, J Appl Physiol, 1996. 81 (6): 2509-16). The control group underwent sham surgery to cut the skin. After surgery, 1 mg / ml ampicillin was added to drinking water. The NOS inhibitor L-NAME (1 mg / ml, Sigma-Aldrich) was administered as drinking water and the drinking water was changed daily. nNOS inhibitor 7-NI (50 mg / kg, Calbiochem) and NOX inhibitor DPI (2 mg / kg, Sigma-Aldrich) were administered intraperitoneally 30 minutes before or 60 minutes after the start of overload. In addition, apocynin (10 mg / kg, Sigma-Aldrich), tadalafil (10 mg / kg, Toronto Research Chemicals), sildenafil (10 mg / kg, Sigma-Aldrich), 8-Br-cGMP (10 mg / kg, Calbiochem ), Rotenone (5 mg / kg, Sigma-Aldrich), allopurinol (10 mg / kg, Cayman), uric acid (200 mg / kg Sigma-Aldrich; Example 2), FeTPPS (30 mg / kg, Calbiochem; Example) 2), ebselen (30 mg / kg, Sigma-Aldrich; Example 2), molsidomine (50 mg / kg, Sigma-Aldrich; Example 2), SIN-1 (10 mg / kg, DOJINDO; Example 2 and 3) SNAP (20 mg / kg, Calbiochem; Example 3) was administered intraperitoneally 30 minutes before the start of overload. BAPTA-AM (50 μM, Calbiochem; Example 3), EGTA (1 mM, Dojindo; Example 3), dantrolene (10 μM, Sigma-Aldrich; Example 3), nifedipine (10 μM, Sigma-Aldrich; Example 3) , BTP2 (10 μM, Calbiochem; Example 3), Gd 3+ (50 μm, Sigma-Aldrich; Example 3), GsMTx-4 (10 μM, Wako; Example 3), thapsigargin (20 μM, Calbiochem; Example 3) Hyp9 (10 μM, Sigma-Aldrich; Example 3) and olvanil (10 μM, Sigma-Aldrich; Example 3) were intramuscularly injected 30 minutes before the start of overload.
安楽死後、足底筋を凍結固定し、8μmに薄切した後にヘマトキシリン・エオジン染色を行った。 After euthanasia, the plantar muscle was frozen and fixed, sliced to 8 μm, and stained with hematoxylin and eosin.
ウエスタンブロットのため、凍結後、薄切した足底筋をサンプルバッファー(0.1%Triton X-100, 50 mM HEPES (pH7.4), 4 mM EGTA, 10 mM EDTA, 15 mM Na4P2O7, 100 mM glycerophosphate, 25 mM NaF, 5 mM Na2VO4, 及びコンプリートプロテアーゼ阻害剤カクテル (Roche))でホモジナイズし、遠心分離(15,000g,10分間)後に上澄を採取した。Coomassie Brilliant Blue G-250(BioRad, CA, USA)を用いてタンパク質濃度を測定した後、等量のサンプルローディングバッファー(30%glycerol, 5%2-mercaptoethanol, 2.3%SDS, 62.5 mM Tris-HCl (pH6.8), 0.05%bromophenol blue)と混ぜ、60℃で15分間の熱変性を行い、30μgをウエスタンブロットに供した。PVDF転写膜はトリス緩衝生理食塩水(TBS)+5%スキムミルク(雪印)でブロッキングを行い、1次抗体を用いて4℃で16時間のインキュベーションを行った。1次抗体にはAkt(#9272, Cell Signaling Technology)、p-Akt(Ser473)(#9271, Cell Signaling Technology)、p70S6K(#9202, Cell Signaling Technology)、及びp-p70S6K(Thr389)(#9205, Cell Signaling Technology)を用いた。転写膜を0.1%Tween 20を含むトリス緩衝生理食塩水(TBST)で洗浄後、2次抗体(ウサギ特異的HRP標識抗体, GE Healthcare)でインキュベートを行い、再度TBSTで洗浄し、ECL Western Blotting Detection System(GE HealthCare, Buckinghamshire, UK)で検出した。
For western blotting, frozen and thinned plantar muscles were added to sample buffer (0.1% Triton X-100, 50 mM HEPES (pH7.4), 4 mM EGTA, 10 mM EDTA, 15 mM Na 4 P 2 O 7 , 100 mM glycerophosphate, 25 mM NaF, 5 mM Na 2 VO 4 , and complete protease inhibitor cocktail (Roche)) and centrifuged (15,000 g, 10 minutes), and the supernatant was collected. After measuring protein concentration using Coomassie Brilliant Blue G-250 (BioRad, CA, USA), an equal volume of sample loading buffer (30% glycerol, 5% 2-mercaptoethanol, 2.3% SDS, 62.5 mM Tris-HCl ( pH 6.8), 0.05% bromophenol blue), heat-denatured at 60 ° C. for 15 minutes, and 30 μg was subjected to Western blotting. The PVDF transfer membrane was blocked with Tris-buffered saline (TBS) + 5% skim milk (snow mark), and incubated for 16 hours at 4 ° C. using the primary antibody. Primary antibodies include Akt (# 9272, Cell Signaling Technology), p-Akt (Ser473) (# 9271, Cell Signaling Technology), p70S6K (# 9202, Cell Signaling Technology), and p-p70S6K (Thr389) (# 9205 , Cell Signaling Technology). After washing the transfer membrane with Tris buffered saline (TBST) containing 0.1
また、NO産生時の副産物であるシトルリン量を測ることで活性測定を行った。凍結後、薄切した足底筋を10倍量の測定バッファー(25 mM Tris-HCl (pH 7.4), 1 mM EDTA, 1 mM EGTA, 及び0.1 mM NaCl)でホモジェナイズし、遠心分離(1,000g,10分間)後に上澄を採取した。30μlの上澄みを60μlのリアクションバッファー([3H]arginine (53.0 Ci/mmol; GE Healthcare), 1 mM NADPH, 750μM CaCl2, 1μM calmodulin, 3μM tetrahydrobiopterin, 3μM flavin-adenine dinucleotide及び3μM flavin mononucleotide)と混ぜ、37℃で20分間インキュベートした。1 mlの反応停止バッファー(2 mM EDTAを含む20 mM HEPES (pH 5.5))を加えた後、AG50WX-8 columns(Na+形態; Dowex)と混ぜ、未反応の[3H]arginineを回収した後、液体シンチレーションカウンターにて[3H]citrulline量を測定した。
In addition, the activity was measured by measuring the amount of citrulline which is a by-product during NO production. After freezing, the thinned plantar muscle is homogenized with 10 times the amount of measurement buffer (25 mM Tris-HCl (pH 7.4), 1 mM EDTA, 1 mM EGTA, and 0.1 mM NaCl) and centrifuged (1,000 g, The supernatant was collected after 10 minutes).
2群間のデータの比較にはstudent-t検定を用いた。また、多群間比較には、ANOVA検定を行った後、Tukey's法による多群間検定を行った。データは平均値±標準誤差を用いて示し、p < 0.05を有意差ありと判定した。 The student-t test was used to compare the data between the two groups. For comparison between multiple groups, ANOVA test was performed and then multiple group test by Tukey's method was performed. Data are shown using mean ± standard error, and p <0.05 was determined to be significant.
同マウスの術後7日の筋重量を計測した結果、野生型は対照群と比べ、過負荷により40%程度筋重量が増加したが、欠損型は20%程度しか増加しなかった(図1のA及びB)。筋肥大におけるNOの寄与を明らかにするため、上記の通りNOS阻害剤であるL-NAMEを投与した。術後3日前から7日後まで、及び術後3日前から1日後までL-NAMEを投与した群は、術後7日の筋重量は有意に低下したが、術後1日以降投与した群では効果は見られなかった(図2)。NO産生がnNOSによるものか検討するため、nNOS特異的阻害剤である7-NIを過負荷30分前若しくは60分後に投与した。その結果、過負荷前に投与した群は有意に筋肥大が抑えられたが、過負荷後に投与した群に効果は見られなかった(図3)。これらの結果から、過負荷開始1時間の間に産生されたnNOSによるNO産生が筋肥大を促進することが示唆された。 As a result of measuring the muscle weight on the 7th day after the operation of the same mouse, the wild type increased the muscle weight by about 40% compared to the control group, but the deficient type increased only about 20% (Fig. 1). A and B). In order to clarify the contribution of NO in muscle hypertrophy, the NOS inhibitor L-NAME was administered as described above. In the group administered L-NAME from 3 days before to 7 days after surgery, and from 3 days before surgery to 1 day after surgery, muscle weight was significantly reduced on the 7th day after surgery, but in the group administered after 1 day after surgery, No effect was seen (Figure 2). To examine whether NO production was due to nNOS, 7-NI, an nNOS-specific inhibitor, was administered 30 minutes before or 60 minutes after overload. As a result, muscle hypertrophy was significantly suppressed in the group administered before overload, but no effect was seen in the group administered after overload (FIG. 3). From these results, it was suggested that NO production by nNOS produced during 1 hour of overload promotes muscle hypertrophy.
過負荷開始3分、1、6及び12時間後にNOS活性測定を行った結果、NOS活性は過負荷開始3分後に上昇し、1時間以降には定常レベル若しくはそれ以下にまで低下した(図4のA)。またこの上昇はnNOS欠損型マウスでは見られなかった(図4のB)。これらの結果から、過負荷直後に活性化されたnNOS及びその後に産生されたNOが筋肥大を促進することが分かった。
As a result of measuring
nNOS野生型では過負荷3分後に、mTORによって制御されるp70S6Kスレオニン389のリン酸化(Zoncu, R., A. Efeyan, and D.M. Sabatini, Nat Rev Mol Cell Biol, 2011. 12(1):21-35)が上昇し、mTORが活性化され、タンパク質合成経路が活性化していたが、欠損型ではその活性化が見られなかった(図5のA及びB)。 In the nNOS wild type, phosphorylation of p70S6K threonine 389 (Zoncu, R., A. Efeyan, and DM Sabatini, Nat Rev Mol Cell Biol, 2011. 12 (1): 21-: 35) increased, mTOR was activated, and the protein synthesis pathway was activated, but the activation was not observed in the defective type (A and B in FIG. 5).
これらの結果から、過負荷直後に活性化したnNOSはmTORによって制御されるタンパク質合成経路の活性化を介して筋肥大を促進することがわかった。 From these results, it was found that nNOS activated immediately after overload promotes muscle hypertrophy through activation of protein synthesis pathway controlled by mTOR.
また、ミトコンドリア呼吸鎖、キサンチンオキシダーゼの阻害剤であるrotenone及びallopurinolを投与しても術後7日の筋重量は低下しなかったが、NOX阻害剤であるapocynin及びDPI投与群は欠損型と同様に筋肥大の進行が抑えられた(図6)。 In addition, administration of rotenone and allopurinol, inhibitors of mitochondrial respiratory chain and xanthine oxidase, did not reduce muscle weight on the 7th postoperative day, but the NOX inhibitor apocynin and DPI administration groups were the same as the deficient type The progression of muscle hypertrophy was suppressed (Fig. 6).
しかし、血管拡張剤であるtadalafil、sildenafil、及びcGMP経路を活性化させる8Br-cGMPを投与しても欠損型の表現型を回復することはできず(データは示さず)、nNOSによって産生されたNOは古典的なNO-cGMP経路(Kemp-Harper, B. and R. Feil, Meeting report: cGMP matters. Sci Signal, 2008. 1(9):pe12)によって制御される血管拡張を介さず、筋肥大を促進することが示唆された。 However, administration of the vasodilators tadalafil, sildenafil, and 8Br-cGMP, which activates the cGMP pathway, did not restore the defective phenotype (data not shown) and were produced by nNOS NO is not mediated by vasodilation controlled by the classical NO-cGMP pathway (Kemp-Harper, B. and R. Feil, Meeting report: cGMP matters. Sci Signal, 2008. 1 (9): pe12) It was suggested to promote hypertrophy.
そこでcGMP非依存的な経路であり、NOとsuperoxideとの反応物であるperoxynitrite(Heck, D.E., Antioxid Redox Signal, 2001. 3(2):249-60)が筋肥大を促進する可能性があると考え、過負荷直後におけるsuperoxide、及びperoxynitriteの機能を明らかにする実験を以下の実施例2及び3で行った。 Therefore, it is a cGMP-independent pathway, and peroxynitrite (Heck, DE, Antioxid Redox Signal, 2001. 3 (2): 249-60), which is a reaction product of NO and superoxide, may promote muscle hypertrophy. Thus, experiments for clarifying the functions of superoxide and peroxynitrite immediately after overload were conducted in Examples 2 and 3 below.
[実施例2]
本実施例では、骨格筋におけるスーパーオキシド(superoxide)の主な供給分子であるミトコンドリア呼吸鎖、キサンチンオキシダーゼ、及びNADPHオキシダーゼ(NOX)の筋肥大における寄与を検討した。
[Example 2]
In this example, the contribution of mitochondrial respiratory chain, xanthine oxidase, and NADPH oxidase (NOX), which are main supply molecules of superoxide in skeletal muscle, to muscle hypertrophy was examined.
過負荷によってNOX活性が上昇するか検討するため、superoxide産生測定を行った。superoxideとルシゲニンとの反応による発光を測定することでsuperoxide産生を測定した。凍結後、薄切した足底筋を測定バッファー(150 mM NaCl, 50 mM Tris-HCl (pH7.4), 25 mM EGTA, 25 mM EDTA, 及びコンプリートプロテアーゼ阻害剤カクテル(Roche))でホモジェナイズし、1,000g、10分間後に上澄を採取した。上澄みを測定バッファー(100 mM KH2PO4, (pH7.0) 及び10μM lucigenin (Sigma-Aldrich))と混ぜた。200μM NADPHは測定直前に反応液に加え、液体シンチレーションカウンターにて測定した。その結果、過負荷直後にsuperoxide産生は上昇しなかった(図7)。 To examine whether NOX activity increases due to overload, superoxide production was measured. Superoxide production was measured by measuring luminescence by reaction between superoxide and lucigenin. After freezing, homogenize sliced plantar muscle with measurement buffer (150 mM NaCl, 50 mM Tris-HCl (pH 7.4), 25 mM EGTA, 25 mM EDTA, and complete protease inhibitor cocktail (Roche)), The supernatant was collected after 1,000 g for 10 minutes. The supernatant was mixed with measurement buffer (100 mM KH 2 PO 4 , (pH 7.0) and 10 μM lucigenin (Sigma-Aldrich)). 200 μM NADPH was added to the reaction solution immediately before the measurement and measured with a liquid scintillation counter. As a result, production of superoxide did not increase immediately after overload (FIG. 7).
また、足底筋におけるNOX1、NOX2、NOX3及びNOX4の発現を、以下に示す配列を有するプライマーセットを用いてRT-PCRにより調べた。 Also, the expression of NOX1, NOX2, NOX3 and NOX4 in the plantar muscle was examined by RT-PCR using a primer set having the following sequence.
足底筋にはNOX2及びNOX4が発現しており(図8)、またNOX4は常時活性化型の酵素(Bedard, K. and K.H. Krause, Physiol Rev, 2007. 87(1):245-313)であることが報告されており、NOX4が筋肥大を促進することが示唆された。 NOX2 and NOX4 are expressed in the plantar muscles (FIG. 8), and NOX4 is a constantly activated enzyme (Bedard, K. and KH Krause, Physiol Rev, 2007. 87 (1): 245-313) It has been reported that NOX4 promotes muscle hypertrophy.
superoxideの筋肥大における機能、及びNOとsuperoxideとの反応物であるペルオキシ亜硝酸(peroxynitrite)の筋肥大における機能を明らかにするため、実施例1に記載のようにsuperoxide及びperoxynitrite消去剤であるebselenと、peroxynitrite消去剤である尿酸、FeTPPSを投与した結果、欠損型と同様に筋肥大の進行が抑えられた(図9)。またperoxynitrite供与剤であるmolsidomine、SIN-1を欠損型に投与した結果、術後7日の筋重量は野生型並みに回復し、この回復はFeTPPSとの共投与により阻害された(図10)。またFeTPPS投与群におけるタンパク質合成経路の活性化を調べた結果、欠損型と同様、過負荷直後においてp70S6Kのリン酸化が見られず、mTORが活性化しなかった(図11のA及びB)。これらの結果から、nNOS由来のNOとNOX由来のsuperoxideから成るperoxynitriteが過負荷直後に産生され、mTORによって制御されるタンパク質合成経路を活性化させることで筋肥大を促進することを見出した。 In order to clarify the function of superoxide in muscle hypertrophy and the function of peroxynitrite, a reaction product of NO and superoxide, in the muscle hypertrophy, as described in Example 1, superoxide and peroxynitrite scavenger ebselen As a result of administration of uric acid and FeTPPS, which are peroxynitrite scavengers, the progression of muscle hypertrophy was suppressed as in the deficient type (FIG. 9). Moreover, as a result of administration of molsidomine and SIN-1 which are peroxynitrite donors to the deficient type, muscle weight on the 7th day after the operation recovered to the same level as the wild type, and this recovery was inhibited by co-administration with FeTPPS (FIG. 10). . In addition, as a result of investigating the activation of the protein synthesis pathway in the FeTPPS administration group, as in the deficient form, phosphorylation of p70S6K was not observed immediately after overload, and mTOR was not activated (A and B in FIG. 11). From these results, it was found that peroxynitrite consisting of nNOS-derived NO and NOX-derived superoxide was produced immediately after overloading, and promoted muscle hypertrophy by activating the protein synthesis pathway controlled by mTOR.
[実施例3]
ペルオキシ亜硝酸(peroxynitrite)は細胞内カルシウム濃度を制御し得る(Trebak, M., et al., Antioxid Redox Signal, 2010. 12(5):657-74)ことから、peroxynitriteが細胞内カルシウム濃度の制御を介してmTORを制御し、筋肥大を促進すると考えた。細胞内カルシウム濃度とmTORの活性化の関係を明らかにするため、筋小胞体によるカルシウムの取り込みを阻害し、細胞内カルシウム濃度を上昇させるthapsigarginを筋注した結果、mTORの活性化が見られた(図12のA及びB)。
[Example 3]
Peroxynitrite can control intracellular calcium concentration (Trebak, M., et al., Antioxid Redox Signal, 2010. 12 (5): 657-74). It was thought that mTOR is controlled via control and promotes muscle hypertrophy. In order to clarify the relationship between intracellular calcium concentration and mTOR activation, intramuscular injection of thapsigargin that inhibits calcium uptake by sarcoplasmic reticulum and raises intracellular calcium concentration resulted in activation of mTOR (A and B in FIG. 12).
細胞内カルシウム濃度と筋肥大の関係を明らかにするため、カルシウムキレート剤であるBAPTA-AM、EGTAを筋注した結果、術後7日の筋重量が有意に抑制された(図13)。また骨格筋において代表的なカルシウムチャネルであるryanodine receptor、L型カルシウムチャネル、TRPチャネルの阻害剤(dantrolene、nifedipine、BTP2)を筋注した。その結果、TRPチャネル阻害薬BTP2によって術後7日の筋重量が有意に抑制されたが、dantrolene、nifedipineによっては抑制されなかった(図13)。また、stretch-activated channelの阻害剤であるGd3+、GsMTx-4を投与しても筋重量が有意に抑制された(図13)。BAPTA-AM及びBTP2は過負荷直後におけるmTORの活性化も抑制したが、dantrolene、nifedipineは効果が見られなかった(図14)。 In order to clarify the relationship between intracellular calcium concentration and muscle hypertrophy, BAPTA-AM and EGTA, which are calcium chelating agents, were injected intramuscularly. As a result, the muscle weight on the 7th day after the operation was significantly suppressed (FIG. 13). In addition, inhibitors of ryanodine receptor, L-type calcium channel, and TRP channel (dantrolene, nifedipine, BTP2), which are typical calcium channels in skeletal muscle, were injected intramuscularly. As a result, the muscle weight on the 7th day after the operation was significantly suppressed by the TRP channel inhibitor BTP2, but not by dantrolene and nifedipine (FIG. 13). In addition, even when Gd 3+ and GsMTx-4, which are inhibitors of the stretch-activated channel, were administered, muscle weight was significantly suppressed (FIG. 13). BAPTA-AM and BTP2 also suppressed mTOR activation immediately after overload, but dantrolene and nifedipine had no effect (FIG. 14).
nNOS欠損型の表現型はNO供与剤(SNAP)、peroxynitrite供与剤(SIN-1)によって回復されるが、その効果はBAPTA-AM、BTP2との共投与により抑制された(図15)。また、欠損型の表現型は、TRPチャネルアゴニスト、すなわちTRPC6チャネルアゴニストであるHyp9(Leuner, K., et al., Mol Pharmacol, 2010. 77(3):368-77)及びTRPV1チャネルアゴニストであるオルバニル(olvanil)の投与により回復された(図16)。 The nNOS deficient phenotype was recovered by NO donor (SNAP) and peroxynitrite donor (SIN-1), but the effect was suppressed by co-administration with BAPTA-AM and BTP2 (FIG. 15). The deficient phenotypes are TRP channel agonists, ie, TRPC6 channel agonists Hyp9 (Leuner, K., et al., Mol Pharmacol, 2010. 77 (3): 368-77) and TRPV1 channel agonists It was recovered by administration of olvanil (FIG. 16).
これらの結果から、過負荷直後に産生されたNO/peroxynitriteはTRPチャネルによる細胞内カルシウム濃度の制御を介して、筋肥大を促進することが明らかとなった(図17)。 These results revealed that NO / peroxynitrite produced immediately after overload promoted muscle hypertrophy through the control of intracellular calcium concentration by TRP channels (FIG. 17).
[参考例1]
筋芽細胞における細胞内カルシウム濃度の変化をin vitroにおいて解析した。具体的には、筋芽細胞株であるC2C12(RCB0987、理研バイオリソースセンターセルバンク)を成長培地(DMEM, 10%ウシ胎児血清, 1%ペニシリン-ストレプトマイシン)で37℃、5%CO2濃度で培養した。その後、分化培地(DMEM, 2%ウマ血清, 1%ペニシリン-ストレプトマイシン)で2日間培養し、筋分化を誘導した。細胞内カルシウム濃度の変化はカルシウム指示薬であるFluo-4(DOJINDO)を用いて計測した。筋分化を誘導したC2C12をPSS溶液(140mM NaCl, 5mM KCl, 2.5mM CaCl2, 1mM MgCl2, 10mM HEPES, 10mM glucose pH7.0)にて6時間以上培養した後、8μMのFluo-4を添加し、30分間室温で培養した。過剰なFluo-4を取り除き、37℃にて5分間培養した後、SIN-1(DOJINDO)による蛍光強度の変化を倒立蛍光顕微鏡(Olympus)を用いて3秒おきに計測した。細胞外カルシウムを取り除く場合、SIN-1を0 Ca2+溶液(140mM NaCl, 5mM KCl, 1mM MgCl2, 10mM HEPES, 10mM glucose, 2mM EGTA pH7.0)を用いて、細胞に添加した。また、SIN-1による処理の際に、peroxynitrite消去剤であるFeTPPS(calbiochem)を100μMの濃度でSIN-1と一緒に添加した。筋小胞体のカルシウムを枯渇させる場合、Fluo-4と共にthapsigargin(Calbiochem, 2μM)を加えた。
[Reference Example 1]
Changes in intracellular calcium concentration in myoblasts were analyzed in vitro. Specifically, myoblast cell line C2C12 (RCB0987, RIKEN BioResource Center Cell Bank) was cultured in growth medium (DMEM, 10% fetal bovine serum, 1% penicillin-streptomycin) at 37 ° C and 5% CO 2 concentration. . Thereafter, the cells were cultured in a differentiation medium (DMEM, 2% horse serum, 1% penicillin-streptomycin) for 2 days to induce muscle differentiation. Changes in intracellular calcium concentration were measured using a calcium indicator Fluo-4 (DOJINDO). C2C12 that induced myogenic differentiation was cultured in PSS solution (140 mM NaCl, 5 mM KCl, 2.5 mM CaCl 2 , 1 mM MgCl 2 , 10 mM HEPES, 10 mM glucose pH 7.0) for 6 hours or more, and then 8 μM Fluo-4 was added. And incubated at room temperature for 30 minutes. After removing excess Fluo-4 and culturing at 37 ° C. for 5 minutes, changes in fluorescence intensity due to SIN-1 (DOJINDO) were measured every 3 seconds using an inverted fluorescence microscope (Olympus). When extracellular calcium was removed, SIN-1 was added to the cells using 0 Ca2 + solution (140 mM NaCl, 5 mM KCl, 1 mM MgCl2, 10 mM HEPES, 10 mM glucose, 2 mM EGTA pH 7.0). In addition, during treatment with SIN-1, FeTPPS (calbiochem), a peroxynitrite scavenger, was added together with SIN-1 at a concentration of 100 μM. To deplete sarcoplasmic reticulum calcium, thapsigargin (Calbiochem, 2 μM) was added along with Fluo-4.
その結果、NO/peroxynitrite供与剤であるSIN-1で細胞を処理した直後から細胞内カルシウム濃度が上昇した(図18A及び図18B)。この細胞内カルシウム濃度の上昇は濃度依存的であり(図18C)、peroxynitrite消去剤であるFeTPPSにより阻害された(図18D)。これらの結果から、peroxynitriteは細胞内カルシウム濃度を上昇させることがわかった。 As a result, the intracellular calcium concentration increased immediately after the cells were treated with SIN-1 which is a NO / peroxynitrite donor (FIGS. 18A and 18B). This increase in intracellular calcium concentration was concentration-dependent (FIG. 18C) and was inhibited by FeTPPS, a peroxynitrite scavenger (FIG. 18D). From these results, it was found that peroxynitrite increases intracellular calcium concentration.
細胞内カルシウム濃度の上昇は、細胞外のカルシウムの取り込み、又は細胞内筋小胞体に貯蔵されているカルシウムの放出によって起こる。細胞外のカルシウムを取り除く(0Ca2+)、又はthapsigarginにより筋小胞体内のカルシウムを枯渇させた。その結果、SIN-1による細胞内カルシウム濃度の上昇は筋小胞体内カルシウムの枯渇によって抑制された(図19A及び図19B)。これらの結果から、peroxynitriteは筋小胞体からのカルシウム放出を誘導することがわかった。 The increase in intracellular calcium concentration occurs due to the uptake of extracellular calcium or the release of calcium stored in the intracellular sarcoplasmic reticulum. Extracellular calcium was removed (0Ca2 +), or calcium in the sarcoplasmic reticulum was depleted by thapsigargin. As a result, the increase in intracellular calcium concentration by SIN-1 was suppressed by depletion of sarcoplasmic reticulum calcium (FIGS. 19A and 19B). These results indicate that peroxynitrite induces calcium release from the sarcoplasmic reticulum.
本明細書中で引用した全ての刊行物、特許及び特許出願は、その全文を参考として本明細書中に取り入れるものとする。 All publications, patents and patent applications cited in this specification are incorporated herein by reference in their entirety.
配列番号1~10:人工配列(合成オリゴヌクレオチド)
Claims (8)
(a)被験物質又は因子による処置を受けた動物から得られた筋細胞において、ペルオキシ亜硝酸及び/又はスーパーオキシド及び/又はカルシウムの濃度を測定するステップ、
(b)(a)の結果に基づいて被験物質又は因子を筋肥大を促進する物質又は因子の候補として同定するステップ
を含む方法。 A method for screening a substance or factor that promotes muscle hypertrophy, comprising:
(A) measuring the concentration of peroxynitrite and / or superoxide and / or calcium in muscle cells obtained from an animal treated with a test substance or factor;
(B) A method comprising identifying a test substance or factor as a candidate substance or factor that promotes muscle hypertrophy based on the result of (a).
(a)筋細胞を、被験物質又は因子により処置するステップ、
(b)該筋細胞において、ペルオキシ亜硝酸及び/又はスーパーオキシド及び/又はカルシウムの濃度を測定するステップ、
(c)(b)の結果に基づいて被験物質又は因子を筋肥大を促進する物質又は因子の候補として同定するステップ
を含む方法。 A method for screening a substance or factor that promotes muscle hypertrophy, comprising:
(A) treating muscle cells with a test substance or factor;
(B) measuring the concentration of peroxynitrite and / or superoxide and / or calcium in the muscle cells;
(C) A method comprising identifying a test substance or factor as a candidate substance or factor that promotes muscle hypertrophy based on the result of (b).
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2011200716 | 2011-09-14 | ||
| JP2011-200716 | 2011-09-14 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2013038740A1 true WO2013038740A1 (en) | 2013-03-21 |
Family
ID=47882986
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2012/057128 Ceased WO2013038740A1 (en) | 2011-09-14 | 2012-03-21 | Method for screening for substance or factor capable of promoting muscle hypertrophy |
Country Status (2)
| Country | Link |
|---|---|
| JP (1) | JPWO2013038740A1 (en) |
| WO (1) | WO2013038740A1 (en) |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2006162346A (en) * | 2004-12-03 | 2006-06-22 | Biomarker Science:Kk | Diagnostic method of amyotrophy, evaluation method of anti-amyotrophic action and screening method of anti-amyotrophic substance |
-
2012
- 2012-03-21 JP JP2013533540A patent/JPWO2013038740A1/en active Pending
- 2012-03-21 WO PCT/JP2012/057128 patent/WO2013038740A1/en not_active Ceased
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2006162346A (en) * | 2004-12-03 | 2006-06-22 | Biomarker Science:Kk | Diagnostic method of amyotrophy, evaluation method of anti-amyotrophic action and screening method of anti-amyotrophic substance |
Non-Patent Citations (11)
| Title |
|---|
| BODINE, S. C. ET AL.: "Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo", NATURE CELL BIOLOGY, vol. 3, no. 11, 2001, pages 1014 - 1019 * |
| ITO, N. ET AL.: "Neuronal nitric oxide synthase (nNOS) is an essential mediator for early stage of muscle hypertrophy", 33RD ANNUAL MEETING OF THE MOLECULAR BIOLOGY SOCIETY OF JAPAN, 83RD ANNUAL MEETING OF THE JAPANESE BIOCHEMICAL SOCIETY GODO TAIKAI KOEN YOSHISHU, 2010 * |
| ITO, N. ET AL.: "Neuronal nitric oxide synthase regulates muscle hypertrophy through Akt signaling pathway", 81ST ANNUAL MEETING OF THE JAPANESE BIOCHEMICAL SOCIETY, 31ST ANNUAL MEETING OF THE MOLECULAR BIOLOGY SOCIETY OF JAPAN GODO TAIKAI KOEN YOSHISHU, 2008 * |
| NIKAWA, T.: "Development of ubiquitin ligase inhibitor as a drug against unloading-mediated muscle atrophy", JOURNAL OF JAPANESE BIOCHEMICAL SOCIETY, vol. 81, no. 7, 2009, pages 614 - 618 * |
| NISHIDA, M. ET AL.: "TRP channels : formation of signal complex and regulation of cellular functions", FOLIA PHARMACOL, vol. 121, 2003, JPN., pages 223 - 232 * |
| NUMATA, T. ET AL.: "Structures and variable functions of TRP channels", JOURNAL OF JAPANESE BIOCHEMICAL SOCIETY, vol. 81, no. 11, 2009, pages 962 - 983 * |
| ROMMEL, C. ET AL.: "Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways", NATURE CELL BIOLOGY, vol. 3, no. 11, 2001, pages 1009 - 1013 * |
| SHIMIZU, N. ET AL.: "Crosstalk between glucocorticoid receptor and nutritional sensor mTOR in skeletal muscle", CELL METABOLISM, vol. 13, February 2011 (2011-02-01), pages 170 - 182, XP028144015, DOI: doi:10.1016/j.cmet.2011.01.001 * |
| SUZUKI, N. ET AL.: "No production results in suspension-induced muscle atrophy through dislocation of neuronal NOS", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 117, no. 9, September 2007 (2007-09-01), pages 2468 - 2476 * |
| TAKEDA, S.: "The role of neuronal nitric oxide synthase (nNOS) in molecular mechanism of muscle atrophy and hypertrophy", 81ST ANNUAL MEETING OF THE JAPANESE BIOCHEMICAL SOCIETY, 31ST ANNUAL MEETING OF THE MOLECULAR BIOLOGY SOCIETY OF JAPAN GODO TAIKAI KOEN YOSHISHU, vol. 4S18-3, 2008 * |
| ZANOU, N. ET AL.: "Role of TRPC1 channel in skeletal muscle function", AM J PHYSIOL CELL PHYSIOL, vol. 298, 2010, pages C149 - C162 * |
Also Published As
| Publication number | Publication date |
|---|---|
| JPWO2013038740A1 (en) | 2015-03-23 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Ma et al. | Leucine and isoleucine have similar effects on reducing lipid accumulation, improving insulin sensitivity and increasing the browning of WAT in high-fat diet-induced obese mice | |
| US10772862B2 (en) | Lipid scavenging in Ras cancers | |
| Li et al. | Berberine improves pressure overload-induced cardiac hypertrophy and dysfunction through enhanced autophagy | |
| Jiang et al. | ATF4 protects against sorafenib-induced cardiotoxicity by suppressing ferroptosis | |
| Chen et al. | Endogenous Nampt upregulation is associated with diabetic nephropathy inflammatory‑fibrosis through the NF‑κB p65 and Sirt1 pathway; NMN alleviates diabetic nephropathy inflammatory‑fibrosis by inhibiting endogenous Nampt | |
| US20170027897A1 (en) | mTORC1 MODULATION BY AMINO ACIDS AND USES THEREOF | |
| US20140011761A1 (en) | Modulating Endoplasmic Reticulum Stress in the Treatment of Tuberous Sclerosis | |
| Peters et al. | Dose-dependent effects of leucine supplementation on preservation of muscle mass in cancer cachectic mice | |
| Gong et al. | Different roles of insulin receptor a and b in maintaining blood glucose homeostasis in zebrafish | |
| Wan et al. | Erythromycin has therapeutic efficacy on muscle fatigue acting specifically on orosomucoid to increase muscle bioenergetics and physiological parameters of endurance | |
| US12472159B2 (en) | Compositions comprising amino acids for use in the prevention and treatment of chemotherapy side effects | |
| US9693994B2 (en) | Class IIa HDAC inhibitors for the treatment of infection | |
| US9072778B2 (en) | Treatment regimen for N-MYC, c-MYC, and L-MYC amplified and overexpressed tumors | |
| Dai et al. | Piperlongumine, a natural alkaloid from Piper longum L. ameliorates metabolic-associated fatty liver disease by antagonizing the thromboxane A2 receptor | |
| WO2013038740A1 (en) | Method for screening for substance or factor capable of promoting muscle hypertrophy | |
| WO2013141202A1 (en) | Muscle-building agent and screening method for muscle-building substance | |
| KR20120058047A (en) | Pharmaceutical composition for preventing and treating obesity comprising sphingosine-1-phosphate | |
| Wang et al. | MEK1/2 Inhibitors Induce Class 1 Alcoholic Dehydrogenase (ADH1) Expression by Regulating Farnesoid X Receptor | |
| RU2558792C2 (en) | Composition for improving general state accompanying hypoalbuminemia | |
| JP2020513232A (en) | Detection method of glucose metabolism abnormality and prevention and treatment | |
| Lee et al. | Muscle extracts from Antarctic fishes Trematomus bernacchii and T. Newnesi enhance myofiber regeneration and muscle function through mTOR signaling | |
| TW202327566A (en) | Use of composition containing fucoxanthin and oligo-fucoidan for preparing pharmaceutical product treating or improving chronic kidney disease, composition containing fucoxanthin, oligo- fucoidan and L-carnitine, and pharmaceutical product containing the same effectively treating or slowing down the chronic kidney disease | |
| Dhanani | Disruption of Branched-Chain Amino Acid Catabolism Impairs Rat Myoblast Survival and Differentiation | |
| Ohyama | Studies on the food compounds showing anti-obesity effect and their mechanism to suppress obesity | |
| Kim | Simultaneously inhibiting multiple nutrient acquisition pathways using synthetic sphingolipid compounds |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 12831023 Country of ref document: EP Kind code of ref document: A1 |
|
| ENP | Entry into the national phase |
Ref document number: 2013533540 Country of ref document: JP Kind code of ref document: A |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 12831023 Country of ref document: EP Kind code of ref document: A1 |