Use of histidine, glycine and other amino acids for the prevention of insulin resistance and/or diabetes

文档序号:602354 发布日期:2021-05-04 浏览:55次 中文

阅读说明:本技术 组氨酸、甘氨酸和其它氨基酸用于预防胰岛素抗性和/或糖尿病的用途 (Use of histidine, glycine and other amino acids for the prevention of insulin resistance and/or diabetes ) 是由 F-P·马丁 J·哈格尔 J·品克内 J·霍斯金 于 2019-09-24 设计创作,主要内容包括:本发明涉及使用至少一种组氨酸或其衍生物、至少一种甘氨酸或其衍生物和至少一种选自N-乙酰基-半胱氨酸、赖氨酸或精氨酸的另外的活性剂的组合的组合物和方法。本发明的一个方面是促进处于发展胰岛素抗性和糖尿病风险的儿童的健康的脂肪代谢和代谢健康,促进健康的脂解以及脂肪酸在代谢中的使用,促进青春期和青少年期期间健康的脂肪和脂肪细胞代谢,治疗或预防氧化应激、与氧化应激相关的病症、降低的谷胱甘肽水平、或与降低的谷胱甘肽水平相关的病症的方法,所述方法通过施用有效量的至少一种甘氨酸或其衍生物与赖氨酸的组合来实现。(The present invention relates to compositions and methods using a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one additional active agent selected from N-acetyl-cysteine, lysine or arginine. One aspect of the invention is a method of promoting healthy fat metabolism and metabolic health in children at risk of developing insulin resistance and diabetes, promoting healthy lipolysis and use of fatty acids in metabolism, promoting healthy fat and adipocyte metabolism during adolescence and adolescence, treating or preventing oxidative stress, disorders related to oxidative stress, reduced glutathione levels, or disorders related to reduced glutathione levels by administering an effective amount of at least one glycine or derivative thereof in combination with lysine.)

1. A composition comprising a combination of at least one histidine, at least one glycine and at least one additional active agent selected from N-acetyl-cysteine, lysine or arginine for use in the prevention of an increase in insulin resistance.

2. A composition comprising a combination of at least one histidine, at least one glycine and at least one additional active agent selected from N-acetyl-cysteine, lysine or arginine for use in (i) preventing an increase in insulin resistance and (ii) preventing or treating diabetes.

3. The composition for the use according to claims 1and 2, comprising a combination of at least one histidine, at least one glycine, and lysine.

4. The composition for use according to claims 1 to 3, wherein the use is for promoting healthy fat metabolism.

5. The composition for use according to any preceding claim, wherein the use is for the treatment or prevention of at least one additional physical condition.

6. The composition for use according to claim 5, wherein the at least one additional physical condition is selected from subcutaneous adipocyte lipolysis, inefficient lipolysis (high basal/low stimulation) or a disorder associated with inefficient lipolysis.

7. The composition for use according to claim 5, wherein the at least one additional body condition is selected from the group consisting of high HOMA-IR, high abdominal glucose and high insulin.

8. The composition for use according to claim 5, wherein the at least one additional physical condition is selected from oxidative stress, a disorder associated with oxidative stress, or a disorder associated with reduced glutathione levels.

9. The composition for use according to claim 5, wherein the at least one additional physical condition is selected from the group consisting of high body weight gain and associated disorders of glucose metabolism during growth and development, high body fat gain and associated disorders of glucose metabolism during growth and development, high center obesity and associated disorders of glucose metabolism during growth and development.

10. The composition for use according to any preceding claim, wherein the use is for enhancing the metabolism of reactive oxygen species, improving glucose control and/or improving muscle function in a subject suffering from at least one of obesity or diabetes.

11. The composition for use according to any preceding claim, wherein the use is for improving mitochondrial function in a subject suffering from sarcopenia.

12. The composition for use according to any preceding claim, wherein the use is by oral administration to an individual in need thereof.

13. A food product comprising a composition, wherein the composition comprises a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one further active agent selected from N-acetyl-cysteine, lysine or arginine, for use in (i) preventing insulin resistance and/or (ii) preventing or treating diabetes.

14. The food product comprising a composition for the use according to claim 13, wherein the composition comprises a combination of at least one histidine, at least one glycine and at least one further active agent selected from N-acetyl-cysteine, lysine or arginine.

15. A kit comprising at least one histidine, at least one glycine and at least one additional active agent selected from lysine, arginine or N-acetyl-cysteine for mixing to form a composition for the use according to claims 1 to 12.

16. The kit of claim 15, wherein the mixing occurs in separate containers as two or more liquid solutions or dry powders.

Detailed Description

The present invention also relates to compositions for promoting metabolic health in an individual at risk of developing insulin resistance.

The present invention also relates to compositions for promoting metabolic health in an individual at risk of developing diabetes.

The present invention also relates to compositions for promoting metabolic health in individuals at risk of developing insulin resistance and diabetes.

The invention also relates to a composition for preventing insulin resistance in an individual.

The invention also relates to a composition for preventing an increase in insulin resistance in an individual.

The present invention also relates to a composition for preventing or treating diabetes in an individual.

The present invention also relates to a composition for use in (i) preventing or preventing an increase in insulin resistance in a subject; and (ii) preventing or treating diabetes in the individual.

In one embodiment, the subject is a human subject. In one embodiment, the human subject is a child. In one embodiment, the human subject is a juvenile. In one embodiment, the human subject is an adult.

The composition comprises at least one histidine or a derivative thereof.

The composition comprises at least one histidine or a derivative thereof, at least one glycine or a derivative thereof, and optionally at least one further active agent selected from N-acetyl-cysteine, lysine, or arginine, or a derivative of said further active agent.

In some embodiments, the composition comprises at least one histidine, at least one glycine and at least one additional active agent selected from N-acetyl-cysteine, lysine or arginine.

In some embodiments, the composition comprises at least one histidine, at least one glycine and at least two additional active agents selected from N-acetyl-cysteine, lysine or arginine.

In one embodiment, the composition comprises at least one histidine, at least one glycine and the additional active agents N-acetyl-cysteine, lysine and arginine.

In some embodiments, the composition is for use in treating or preventing at least one additional physical condition as described herein.

In some embodiments, the composition is for use in treating or preventing at least one additional medical condition, wherein the medical condition is an inflammatory disease. In one embodiment, the inflammatory disease is treated or prevented in a juvenile male. In one embodiment, the adolescent male is 13 years old or 14 years old. In one embodiment, the composition comprises at least one histidine or derivative thereof, at least one glycine or derivative, and optionally at least one lysine.

In some embodiments, the composition comprises a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof, and at least one lysine or derivative thereof for use in the treatment or prevention of at least one physical condition selected from the group consisting of: inefficient lipolysis, such as high basal lipolysis, low stimulated lipolysis, or a condition associated with inefficient lipolysis.

In some embodiments, the composition comprises a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and lysine for promoting and maintaining effective subcutaneous fat cell lipolysis and fatty acid metabolism.

In some embodiments, the composition comprises a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and lysine for use in treating or preventing at least one physical condition selected from the group consisting of high HOMA-IR, high abdominal glucose and high insulin.

In some embodiments, the composition comprises a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and lysine for use in the treatment or prevention of at least one condition selected from oxidative stress, a disorder associated with oxidative stress, or a disorder associated with reduced glutathione levels.

In some embodiments, the composition comprises a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and lysine for use in the treatment or prevention of at least one physical condition selected from the group consisting of high body weight gain and associated disorders of glucose metabolism during growth and development, high body fat gain and associated disorders of glucose metabolism during growth and development, high center obesity and associated disorders of glucose metabolism during growth and development.

In another embodiment, the composition comprises a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one lysine or a derivative thereof for use in enhancing the metabolism of reactive oxygen species, improving glucose control and/or improving muscle function in an individual suffering from at least one of obesity or diabetes.

In another embodiment, the composition comprises a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one arginine or a derivative thereof for use in enhancing the metabolism of reactive oxygen species, improving glucose control and/or improving muscle function in an individual suffering from at least one of obesity or diabetes.

In another embodiment, the composition comprises a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one N-acetyl-cysteine or a derivative thereof for use in enhancing the metabolism of reactive oxygen species, improving glucose control and/or improving muscle function in an individual suffering from at least one of obesity or diabetes.

In another embodiment, the composition comprises a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one lysine or a derivative thereof for use in improving mitochondrial function in a subject suffering from sarcopenia. An individual suffering from sarcopenia may be otherwise healthy or sarcopenia.

In another embodiment, the composition comprises a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one arginine or derivative thereof for use in improving mitochondrial function in an individual with sarcopenia. An individual suffering from sarcopenia may be otherwise healthy or sarcopenia.

In another embodiment, the composition comprises a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one N-acetyl-cysteine or a derivative thereof for use in improving mitochondrial function in an individual suffering from sarcopenia. An individual suffering from sarcopenia may be otherwise healthy or sarcopenia.

In one embodiment, the composition of the invention is used for the treatment or prevention of at least one deleteriously affected condition selected from the group consisting of: type I diabetes, type II diabetes, diabetic complications, insulin resistance, metabolic syndrome, dyslipidemia, overweight, obesity, elevated cholesterol levels, elevated triglyceride levels, elevated fatty acid levels, fatty liver disease, cardiovascular disease, myopathy such as statin induced myopathy, non-alcoholic steatohepatitis, hypertension, atherosclerosis/coronary artery disease, post-stress myocardial injury.

In one embodiment, the composition is a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one further active agent selected from N-acetyl-cysteine, lysine or arginine for use via oral administration according to the present invention.

In one embodiment, the composition is a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one further active agent selected from N-acetyl-cysteine, lysine or arginine for use in administering in a food product according to the present invention.

In one embodiment, the composition is a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and N-acetyl-cysteine or functional derivative thereof for use according to the present invention, wherein the dipeptide provides at least a portion of the at least one glycine or functional derivative thereof and the at least one N-acetyl cysteine or functional derivative thereof.

Each of these compounds may be administered simultaneously (i.e., as a single unit) with the other compounds or separately (i.e., in separate units) at certain intervals.

In one embodiment, the composition is a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one lysine or derivative thereof used according to the present invention via administration as a single unit in the same composition.

In one embodiment, the composition is a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one arginine or derivative thereof for use in accordance with the present invention via administration as a single unit in the same composition.

In one embodiment, the composition is a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one N-acetyl cysteine or derivative thereof for use via administration as a single unit in the same composition according to the present invention.

In one embodiment, the composition is a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one lysine or derivative thereof for use via administration in separate units according to the present invention.

In one embodiment, the composition is a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one arginine or derivative thereof for use via administration in separate units according to the present invention.

In one embodiment, the composition is a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one N-acetyl cysteine or derivative thereof for use via administration in separate units according to the present invention.

The compositions of the present invention comprise the combination in an amount effective for at least one of: (i) subcutaneous adipocyte lipolysis and use of fatty acids in metabolism, inefficient lipolysis (high basal/low stimulation), conditions associated with inefficient lipolysis, (ii) high HOMA-IR, high abdominal glucose and insulin, (iii) oxidative stress, conditions associated with oxidative stress or conditions associated with reduced glutathione levels, (iv) high body weight gain during growth and development and associated glucose metabolism disorders, high body fat gain during growth and development and associated glucose metabolism disorders, high central obesity during growth and development and associated glucose metabolism disorders.

In some embodiments, the compositions of the present invention comprise the combination in an amount effective for at least one of: (i) disorders associated with inefficient lipolysis, (ii) disorders associated with high IR, (iii) disorders associated with oxidative stress, (iv) disorders associated with high body weight gain during growth and development and associated disturbances in glucose metabolism.

In one embodiment, the composition of the invention is a food product for use according to the invention.

The invention also relates to a kit comprising at least one histidine or a derivative thereof, at least one glycine or a derivative thereof, and at least one of the following amino acids: lysine, arginine or N-acetyl-cysteine, or derivatives thereof, for mixing to form one or more compositions disclosed herein and/or for use according to the invention, e.g. in separate containers as two or more liquid solutions or dry powders. In some embodiments, one or more of these compounds may be an isolated compound.

The combination of at least one glycine or derivative thereof and at least one N-acetyl cysteine or functional derivative thereof may be provided by any of the compositions disclosed below: U.S. Pat. Nos. 8,362,080, 8,802,730 and 9,084,760 (each entitled "influencing Glutathione Levels for therapy") and WO2016/191468 (entitled "cosmetics of Supplementation with N-acetyl cysteine and Glycine to improving Glutathione Levels"), each of which is hereby incorporated by reference in its entirety.

Accordingly, one aspect of the present invention is a composition comprising at least one histidine or a derivative thereof, at least one glycine or a derivative thereof, and at least one of the following amino acids: lysine, arginine or N-acetyl-cysteine, or a derivative thereof, in an amount effective for treating or preventing a condition of deleterious effect selected from at least: (i) subcutaneous adipocyte lipolysis and use of fatty acids in metabolism, inefficient lipolysis (high basal/low stimulation), conditions associated with inefficient lipolysis, or (ii) high HOMA-IR, high abdominal glucose and insulin, or (iii) oxidative stress, conditions associated with oxidative stress, or conditions associated with decreased glutathione levels, or (iv) high body weight gain during growth and development and associated glucose metabolism disorders, high body fat gain during growth and development and associated glucose metabolism disorders, high central obesity during growth and development and associated glucose metabolism disorders.

The present invention also relates generally to methods for promoting metabolic health, particularly in individuals at risk of developing insulin resistance and/or diabetes.

The invention also relates to methods for preventing or preventing an increase in insulin resistance.

The invention also relates to methods for preventing or treating diabetes.

The present invention also relates to a method for (i) preventing or preventing an increase in insulin resistance; and (ii) preventing or treating diabetes.

In one embodiment, the subject is a human subject. In one embodiment, the human subject is a child. In one embodiment, the human subject is a juvenile. In one embodiment, the human subject is an adult.

The present invention also relates generally to methods for promoting healthy fat metabolism in a human subject at risk of developing insulin resistance and diabetes.

The method of the invention comprises administering a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and optionally at least one additional active agent selected from N-acetyl-cysteine, lysine or arginine.

In some embodiments, the method is for treating or preventing inefficient lipolysis, such as high basal lipolysis, low stimulated lipolysis, or a condition associated with inefficient lipolysis, the method comprising administering an effective amount of a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof, and lysine.

In some embodiments, a method for promoting healthy fat metabolism comprises promoting and maintaining healthy subcutaneous fat cell lipolysis and fatty acid metabolism, the method comprising administering an effective amount of a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof, and lysine.

In some embodiments, the method is for treating or preventing at least one physical condition selected from the group consisting of high HOMA-IR, high fasting glucose and high insulin, said method comprising administering an effective amount of a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and lysine.

In some embodiments, the method is for treating or preventing at least one condition selected from oxidative stress, a disorder associated with oxidative stress, or a disorder associated with reduced levels of glutathione, the method comprising administering an effective amount of a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof, and lysine.

In some embodiments, the method is for treating or preventing at least one condition selected from the group consisting of high body weight gain and associated disorders of glucose metabolism during growth and development, high body fat gain and associated disorders of glucose metabolism during growth and development, high center obesity and associated disorders of glucose metabolism during growth and development, comprising administering an effective amount of a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and lysine.

In another embodiment, the method is for enhancing the metabolism of reactive oxygen species, improving glucose control, and/or improving muscle function in an individual having at least one of obesity or diabetes, the method comprising administering to the individual an effective amount of a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof, and at least one lysine or derivative thereof.

In another embodiment, the method is for enhancing the metabolism of reactive oxygen species, improving glucose control, and/or improving muscle function in an individual having at least one of obesity or diabetes, the method comprising administering to the individual an effective amount of a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof, and at least one arginine or derivative thereof.

In another embodiment, the method is for enhancing the metabolism of reactive oxygen species, improving glucose control, and/or improving muscle function in a subject suffering from at least one of obesity or diabetes, the method comprising administering to the subject an effective amount of a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof, and at least one N-acetyl-cysteine or derivative thereof.

In another embodiment, the method is for improving mitochondrial function in a subject suffering from sarcopenia, the method comprising administering to the subject an effective amount of a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one lysine or a derivative thereof. An individual suffering from sarcopenia may be otherwise healthy or sarcopenia.

In another embodiment, the method is for improving mitochondrial function in an individual having sarcopenia comprising administering to the individual an effective amount of a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one arginine or derivative thereof. An individual suffering from sarcopenia may be otherwise healthy or sarcopenia.

In another embodiment, the method is for improving mitochondrial function in an individual suffering from sarcopenia, the method comprising administering to the individual an effective amount of a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one N-acetyl-cysteine or derivative thereof. An individual suffering from sarcopenia may be otherwise healthy or sarcopenia.

In one embodiment, the method is for treating or preventing at least one deleteriously affected medical condition selected from the group consisting of: type I diabetes, type II diabetes, diabetic complications, insulin resistance, metabolic syndrome, dyslipidemia, overweight, obesity, elevated cholesterol levels, elevated triglyceride levels, elevated fatty acid levels, fatty liver disease, cardiovascular disease, myopathy such as statin induced myopathy, non-alcoholic steatohepatitis, hypertension, atherosclerosis/coronary artery disease, post-stress myocardial injury.

In one embodiment, the at least one glycine or derivative thereof is selected from the group consisting of L-glycine, L-glycine ethyl ester, D-allylglycine; n- [ bis (methylthio) methylene ] glycine methyl ester; Boc-allyl-Gly-OH (dicyclohexylammonium) salt; Boc-D-Chg-OH; Boc-Chg-OH; (R) -N-Boc- (2' -chlorophenyl) glycine; Boc-L-cyclopropylglycine; Boc-L-cyclopropylglycine; (R) -N-Boc-4-fluorophenylglycine; Boc-D-propargylglycine; boc- (S) -3-thienylglycine; boc- (R) -3-thienylglycine; d-a-cyclohexylglycine; l-a-cyclopropylglycine; n- (2-fluorophenyl) -N- (methylsulfonyl) glycine; n- (4-fluorophenyl) -N- (methylsulfonyl) glycine; Fmoc-N- (2, 4-dimethoxybenzyl) -Gly-OH; n- (2-furoyl) glycine; l-a-neopentylglycine; d-propargylglycine; sarcosine; trimethyl Z-a-phosphonoglycine and mixtures thereof.

In one embodiment, a combination of at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one additional active agent such as N-acetyl-cysteine, lysine or arginine is administered orally.

In one embodiment, a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one additional active agent such as N-acetyl-cysteine, lysine or arginine is administered in a food product.

In one embodiment, a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof, and at least one additional active agent such as N-acetyl-cysteine, lysine, or arginine is administered in a composition comprising a dipeptide that provides at least a portion of the at least one glycine or a derivative thereof and the at least one N-acetyl cysteine or a derivative thereof.

In one embodiment, at least one histidine or functional derivative thereof, at least one glycine or derivative thereof and at least one lysine or derivative thereof are administered in the same composition.

In one embodiment, at least one histidine or functional derivative thereof, at least one glycine or derivative thereof and at least one arginine or derivative thereof are administered in the same composition.

In one embodiment, at least one histidine or derivative thereof, at least one glycine or derivative thereof and at least one N-acetyl cysteine or derivative thereof are administered in the same composition.

In one embodiment, at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one lysine or a derivative thereof are administered in a different composition relative to the remainder of the combination.

In one embodiment, at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one arginine or a derivative thereof are administered in a different composition relative to the remainder of the combination.

In one embodiment, at least one histidine or a derivative thereof, at least one glycine or a derivative thereof and at least one N-acetyl cysteine or a derivative thereof are administered in a different composition relative to the remainder of the combination.

By "weight management" of an adult (e.g., at least eighteen years of age from birth) is meant that the individual has a Body Mass Index (BMI) of about the same after one week, preferably one month, more preferably one year, of consumption of the composition relative to its BMI at the start of consumption of the composition. By "weight management" of a younger individual is meant that after one week of consumption of the composition, preferably after one month of consumption of the composition, more preferably after one year of consumption of the composition, the BMI of the individual for the corresponding age is about the same percentile relative to the BMI percentile at the beginning of consumption of the composition.

In a related embodiment, a method of weight management of an individual comprises administering to the individual a composition comprising an effective amount of a combination of at least one histidine or a derivative thereof, at least one glycine or a derivative thereof, and at least one of the following amino acids: lysine, arginine or N-acetyl-cysteine or derivatives thereof.

For example, a peptide comprising at least one histidine or derivative thereof, at least one glycine or derivative thereof, and at least one of the following amino acids: compositions of combinations of lysine, arginine, or N-acetyl-cysteine or derivatives thereof are administered to individuals who are managing their weight or are on a weight loss program. The weight loss program may include, for example, a weight loss diet (e.g., one or more of: a low fat diet, e.g., a diet less than 20% of the calories from fat, preferably less than 15% of the calories from fat, a low carbohydrate diet, e.g., a diet less than 20% of the calories from carbohydrates, a low calorie diet, e.g., a diet with less calories per day relative to the individual's prior meal intake, or a diet with less calories per day relative to an average person of similar size, or a very low calorie diet, e.g., a diet of 800kcal (3,300kJ) or less per day). Additionally or alternatively, the weight loss program can include a weight loss training regimen (e.g., endurance training and/or strength training).

The method may comprise identifying an individual in need of weight management or weight loss and/or identifying an individual who is obese or overweight, for example prior to initial administration of the composition.

In each of the compositions and methods disclosed herein, the composition is preferably a food product, including a food additive, food ingredient, functional food, dietary supplement, medical food, nutraceutical, or food supplement.

Histidine is an amino acid with the chemical name 2-amino-3- (3H-imidazol-4-yl) propionic acid. Histidine exists in two enantiomeric forms, L-histidine and D-histidine, as shown below:

reference herein to the general term "histidine" includes any scalemic or racemic mixture of enantiomers (wherein a scalemic mixture comprises enantiomers in any relative proportion and a racemic mixture comprises enantiomers in a ratio of 50: 50) as well as L-histidine and/or D-histidine. References herein to an individual enantiomer are specific to that enantiomer only. When providing a scalemic mixture, the mixture preferably comprises more L-histidine than D-histidine, more preferably the mixture comprises predominantly L-histidine. For example, the scalemic mixture may comprise at least 60%, more preferably at least 75%, even more preferably at least 90% by weight of L-histidine. The generic term "histidine" as referred to herein also includes all tautomeric forms. Preferably, the histidine is L-histidine and/or a derivative thereof. L-histidine occurs naturally and is readily available from natural sources.

Histidine can be synthesized from suitable starting materials using standard procedures of organic chemistry, or can be isolated from natural sources using well-known procedures. The synthesis or isolation of a particular enantiomer of histidine may be carried out by standard techniques of organic chemistry well known in the art, for example by synthesis from optically active starting materials or by resolution of the racemic form (for example by suitable well known techniques).

L-histidine can be isolated, for example, from any suitable source, such as meat, poultry, dairy, fish, rice, wheat, and rye.

Histidine may be provided as a solid or semi-solid, preferably as a powder.

Any suitable derivative of histidine may be used in the present invention, provided that the derivative is suitable for inclusion in a pharmaceutical composition and provides the desired pharmacological effect as discussed herein. Combinations of histidine and suitable derivatives thereof may be used. Derivatives of histidine may be synthesized from the appropriate starting materials using standard procedures of organic chemistry, or may be isolated from natural sources using well-known procedures.

Suitable derivatives of histidine may comprise predominantly a histidine core, with minor modifications to the functional groups of the histidine core. For example, reference herein to derivatives of histidine includes compounds derived from histidine (i.e., having a histidine core), wherein the carboxylic acid or amino group of the histidine core is derivatized to include substituents or alternative functional groups. For example, suitable derivatives in which the hydroxyl group of the carboxylic acid group is derivatized may include esters (such as esters formed by reacting a carboxylic acid with an alcohol (such as methanol, ethanol, isopropanol, or butanol)). Suitable derivatives in which the amino group is derivatized may include dialkylamines or trialkylamines (such as those formed by reaction of an amino group with an alkyl halide).

Further suitable derivatives of histidine include peptides of histidine, such as peptides comprising 2 or more histidine units, for example 2 to 20 histidine units, in particular 2 to 10 histidine units or 7 to 10 histidine units, or for example 20 or more histidine units. Particular such derivatives may be di-and tripeptides of histidine.

Further suitable derivatives of histidine include peptides of histidine and one or more further amino acids, such as peptides comprising 2 or more histidines/further amino acid units, for example 2 to 20 histidines/further amino acid units, in particular 2 to 10 histidines/further amino acid units or 7 to 10 histidines/further amino acid units, or for example 20 or more histidines/further amino acid units. Particular such derivatives may be di-and tripeptides, such as di-and beta-alanine dipeptides (also known as carnosine).

Additional suitable derivatives may include, for example, pharmaceutically acceptable salts or prodrugs of histidine and functionalized compounds or polypeptides as described above. By the term prodrug is meant a compound that decomposes in an individual, for example in a warm blooded animal such as man, to release histidine and/or a derivative thereof. Examples of prodrugs may include in vivo cleavable ester derivatives such as those described above. Suitable pharmaceutically acceptable salts and prodrugs are based on sound medical judgment as being suitable for administration to an individual, e.g. a warm-blooded animal such as man, without undesirable pharmacological activity and without undue toxicity. Examples of suitable pharmaceutically acceptable salts include acid addition salts formed with inorganic or organic acids such as hydrochloric, hydrobromic, sulfuric, trifluoroacetic, citric or maleic acid.

In all aspects of the invention as described herein, any derivative of histidine is preferably selected from one or more of peptides of histidine (particularly di-and tripeptides of histidine), peptides of histidine and one or more additional amino acids (particularly di-and tripeptides, e.g. carnosine), and pharmaceutically acceptable salts of histidine. More preferably, any derivative of histidine is selected from one or more of a dipeptide or tripeptide of histidine, a dipeptide or tripeptide of histidine and one or more additional amino acids (e.g. carnosine), and a pharmaceutically acceptable salt of histidine. Even more preferably, any derivative of histidine is a pharmaceutically acceptable salt of histidine.

Most preferably, in all aspects of the invention as described herein, the active ingredient is histidine, more preferably L-histidine. In other words, the present invention preferably provides histidine (more preferably L-histidine) as described herein for use in maintaining and/or improving the barrier function of the skin of an individual, and for use in preventing a skin disorder (especially an inflammatory skin disease, more particularly a chronic inflammatory skin disease, even more particularly atopic dermatitis) as described herein, as well as pharmaceutical compositions and nutritional products comprising histidine (more preferably L-histidine) as described herein.

Lysine, its isomeric forms (L-or D-, alone or in various combinations between them), its salts and short oligomers (most preferably up to m.w.1000) and its salts, derivatives (e.g. acetyl lysine/oligo-lysine) as active ingredient (with or without one or more additives) may be used according to the invention.

The glycine is preferably L-glycine and/or L-glycine ethyl ester. Non-limiting examples of suitable glycine functional derivatives include D-allyl glycine; n- [ bis (methylthio) methylene ] glycine methyl ester; Boc-allyl-Gly-OH (dicyclohexylammonium) salt; Boc-D-Chg-OH; Boc-Chg-OH; (R) -N-Boc- (2' -chlorophenyl) glycine; Boc-L-cyclopropylglycine; Boc-L-cyclopropylglycine; (R) -N-Boc-4-fluorophenylglycine; Boc-D-propargylglycine; boc- (S) -3-thienylglycine; boc- (R) -3-thienylglycine; d-a-cyclohexylglycine; l-a-cyclopropylglycine; n- (2-fluorophenyl) -N- (methylsulfonyl) glycine; n- (4-fluorophenyl) -N- (methylsulfonyl) glycine; Fmoc-N- (2, 4-dimethoxybenzyl) -Gly-OH; n- (2-furoyl) glycine; l-a-neopentylglycine; d-propargylglycine; sarcosine; trimethyl Z-a-phosphonoglycine and mixtures thereof.

In one embodiment, both glycine and N-acetylcysteine may be provided in the form of a dipeptide, such as N-acetylcysteinyl glycine or cysteinyl glycine.

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

N-acetylcysteine or a functional derivative thereof may be administered in an amount of about 0.1 to 100 milligrams (mg) of N-acetylcysteine or a functional derivative thereof per kilogram (kg) of the subject's body weight. Glycine or a functional derivative thereof may be administered in an amount of about 0.1 to 100 milligrams (mg) glycine or a functional derivative thereof per kilogram (kg) of the body weight of the subject.

The compositions disclosed herein can be administered to an individual orally or parenterally. Non-limiting examples of parenteral administration include intravenous, intramuscular, intraperitoneal, subcutaneous, intra-articular, intrasynovial, intraocular, intrathecal, topical and inhalation. Thus, non-limiting examples of composition forms include natural foods, processed foods, natural juices, concentrates and extracts, injectable solutions, microcapsules, nanocapsules, liposomes, ointments, inhalation forms, nasal sprays, nasal drops, eye drops, sublingual tablets, and sustained release formulations.

The compositions disclosed herein can be administered therapeutically using any of a variety of formulations. More specifically, the pharmaceutical composition may comprise a suitable pharmaceutically acceptable carrier or diluent, and may be formulated into preparations in the form of solid, semisolid, liquid or gas, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres and aerosols. Thus, administration of the composition can be accomplished in a variety of ways, including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, and intratracheal administration. The active agent may be systemic after administration, or may be localized through the use of local administration, intramural administration, or the use of an implant that acts to maintain the active dose at the site of implantation.

In pharmaceutical dosage forms, the compounds may be administered as their pharmaceutically acceptable salts. They may also be used in appropriate combination with other pharmaceutically active compounds. The following methods and excipients are exemplary only, and not in any way limiting.

For oral formulations, the compounds can be used alone or in combination with suitable additives for the preparation of tablets, powders, granules or capsules, for example in combination with conventional additives, such as lactose, mannitol, corn starch or potato starch; in combination with a binder, such as crystalline cellulose, a functional derivative of cellulose, gum arabic, corn starch or gelatin; in combination with a disintegrant such as corn starch, potato starch or sodium carboxymethyl cellulose; in combination with a lubricant, such as talc or magnesium stearate; and if desired, in combination with diluents, buffers, wetting agents, preservatives and flavouring agents.

The compounds may be formulated for injection by: dissolving, suspending or emulsifying these compounds in an aqueous or non-aqueous solvent (such as vegetable oil or other similar oils, synthetic aliphatic glycerides, esters of higher aliphatic acids or propylene glycol); and these compounds are used together with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifiers, stabilizers and preservatives, if necessary.

These compounds are useful in aerosol formulations to be administered by inhalation. For example, the compounds may be formulated as pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.

In addition, these compounds can be formulated into suppositories by mixing with various bases such as emulsifying bases or water-soluble bases. The compounds may be administered rectally by means of suppositories. Suppositories may contain vehicles such as cocoa butter, carbowax (carbowax) and polyethylene glycol, which melt at body temperature but solidify at room temperature.

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

It should be understood that various changes and modifications to the presently preferred embodiments described herein will be apparent to those skilled in the art. Such changes and modifications can be made without departing from the spirit and scope of the present subject matter and without diminishing its intended advantages. Accordingly, such changes and modifications are intended to be covered by the appended claims.

In one aspect of the invention, the change in diet is associated with athletic activity program management. The physical activity program should be adapted to the individual's body composition, medical condition and age, with the aim of reducing or managing body weight, and improving body fat mass and lean mass to obtain optimal glucose management results.

For example, the solution may be part of a sporting activity program that uses all of the students' opportunities to perform sporting activities that meet the national recommendations of minutes of sporting activity per day (e.g., moderate to severe sporting activities of 60 minutes per day). For example, the program may follow public health guidelines for physical activity of children and adolescents (e.g., the uk national institute for health and wellness:https://www.nice.org.uk/guidance)。

examples

Example 1

Methods used in the study

Study population

The EarlyBird diabetes study included a cohort born at 1995/1996 and was recruited in 2000/2001 years when the children were 5 years old (307 children, 170 boys). The data set from the EarlyBird cohort consists of several clinical and anthropometric variables measured annually from 5 to 16 years of age. The study was performed according to DE-Dow guidelines of the second Declaration of Helsinki II of Helsinki; DE approval was granted by the Plymous Local Research DE Committee (Plymouth Local Research Ethics Committee) (1999), with written consent from parents and oral consent from children.

Tracking good cohort retention at the age when some young people will start moving from home to their own lives. Follow-up studies were prepared starting at 6 months in 2013 and study visits started at 2 months in 2015 until 2016 summer. A total of 178 Earlybird participants completed this follow-up as adults (average age 20 years), with data collected using an adapted study protocol.

Anthropometric parameters

BMI was derived from direct measurements of height (Leicester height measurement; Child Growth Foundation, London, U.K.) and body weight (Tanita Solar 1632 scale), performed in a blind and repeated manner and averaged. BMI SD scores were calculated according to the british 1990 standard.

Athletic activity measurements were performed annually from 5 years old using an accelerometer (Acti-Graph [ formerly MTI/CSA ]). The child is required to wear the accelerometer continuously for 7 days at each annual time point and only use the recordings captured for at least 4 days.

The resting energy consumption was measured by indirect calorimetry using the vent flow hood technique (gas exchange measurement, Manchester Nutren technologies Ltd, Manchester, UK). Performance tests are reported to show an average error in measuring oxygen consumption of 0.3 ± 2.0%, and an average error in carbon dioxide production of 1.8 ± 1%. Measurements were performed in a quiet hot neutral chamber (20 ℃) after an overnight fast period of at least 6 hours to minimize any effects due to thermal effects of food. Data was collected for at least 10 minutes and Respiratory Quotient (RQ) was calculated as an indicator of Basal Metabolic Rate (BMR).

Clinical parameters

After an overnight fasting period, peripheral blood was collected into EDTA tubes each year and stored at-80 ℃. Insulin Resistance (IR) was determined annually using the homeostatic model evaluation program (HOMA-IR), which has been validated in children, from fasting plasma glucose (Cobas Integr 700 Analyzer; Roche Diagnostics) and insulin (DPC IMMULITE) (cross-reactivity with proinsulin, 1%).

Serum metabolomics

Mu.l of serum was mixed with 200. mu.l of deuterated phosphate buffer solution 0.6M KH2PO4 containing 1mM of 3- (trimethylsilyl) - [2,2,3,3-2H4] -1-propionic acid sodium (TSP, chemical shift reference δ H ═ 0.0 ppm). Transfer 550 μ L of the mixture to a 5mm NMR tube.

1H NMR metabolic spectra of serum samples were obtained with a Bruker Avance III 600MHz spectrometer (Bruker Biospin, Rheinstetten, Germany) equipped with a 5mm cryoprobe and at a temperature of 310K, and processed using the TOPSPIN (version 2.1, Bruker Biospin, Rheinstetten, Germany) software package, as previously described. Standards were obtained using 32 scans and 98000 data points1H NMR one-dimensional pulse sequence (with water suppression), Carr-Purcell-Meiboom-gill (cpmg) spin echo sequence (with water suppression), and diffusion editing sequence. The spectral data (from δ 0.2 to δ 10) were imported into Matlab software (version R2013b, meiswoks Inc (Mathworks Inc, Natwick MA), na tik, usa) with a resolution of 22000 data points and normalized to total area after removal of the solvent peaks. Poor quality or highly diluted spectra were discarded from subsequent analyses.

The 1H-NMR spectrum of human plasma enables monitoring of the signals associated with the fatty acyl groups bound to lipoproteins present in triglycerides, phospholipids and cholesterol esters, as well as the peaks from the glyceryl of triglycerides and the choline head group of phosphatidylcholine. This data also covers the quantitative analysis of the major low molecular weight molecules present in blood. Based on internal databases, representative signals for 1H CPMG NMR spectrally assignable metabolites are integrated, including asparagine, leucine, isoleucine, valine, 2-ketobutyric acid, 3-methyl-2-oxoacetic acid, α -ketoisovaleric acid, (R) -3-hydroxybutyric acid, lactic acid, alanine, arginine, lysine, acetic acid, N-acetylglucosamine, O-acetylglucosamine, acetoacetic acid, glutamic acid, glutamine, citric acid, dimethylglycine, creatine, citrulline, trimethylamine N-oxide, taurine, proline, methanol, glycine, serine, creatinine, histidine, tyrosine, formic acid, phenylalanine, threonine, and glucose. Furthermore, in diffusion-edited spectra, signals associated with different lipid classes were integrated, including phospholipids containing choline, VLDL isoforms, unsaturated fatty acids, and polyunsaturated fatty acids. The signal is expressed in arbitrary units corresponding to peak areas normalized to the total metabolic profile, which represents the relative change in the metabolite concentration in serum.

Mass spectrometry-based determination of serum amino acids

Serum amino acid quantification of selected samples was performed using an internal automated quantification method of amino acids in human plasma and serum by UPLC-MS/MS. Briefly, after the precipitation, derivatization and dilution steps, the samples were subjected to liquid chromatography (Acquity class I, Waters) coupled with mass spectrometry analysis (Xevo TQ-XS triple quadrupole, Waters). For chromatographic separation, the gradient consisted of a mobile phase of ammonium formate (0.55g/L aqueous ammonium formate solution, 0.1% formic acid) and a second mobile phase of acetonation (acetonitrile 0.1% formic acid). The analyte concentration was calculated from the peak area ratio of the compound to its corresponding internal standard. The results are expressed in μ M. Peaks were integrated using the AA _ quantitionemeth in TargetLynx function included in MassLynx software.

Statistical analysis

The distribution of the resulting variable IR is skewed and therefore logarithmically transformed for analysis. For both experimental and primary study analyses, data at all ages were used simultaneously, and mixed effect modeling was used to assess the association between IR (HOMA-IR) and individual metabolites, taking into account age, BMI sds, physical activity, and adolescence schedule (APHV). Including random truncation as well as age (classified to allow non-linear changes in IR over time), gender, DEXA% fat, APHV, MVPA (minutes of moderate to severe physical activity) and individual metabolites (in separate models) as fixed effects.

The inventors performed a first study (experimental study) on a subset of 40 participants aged 5 to 14 years and evaluated the reproducibility of another subset of 150 participants aged 5 to 16 years (main study). In the experimental study, 40 individuals were selected based on having a complete set of samples available for analysis (stratified by IR at ages 5 and 14) at each time point between 5 and 14 years (20 boys). In the main study, 150 individuals were selected to include all individuals who had shown impaired fasting glucose at one or more time points during the study. Only 28 children were common to both studies. Individuals showing fasting glucose abnormality were gender matched, thereby selecting 105 boys and 45 girls.

To further assess which IR-related metabolites may be early indicators of IR trajectories, the inventors stratified the main study population according to low or high IR states in the 14-16 year old range. Optionally, the 91 th percentile of the HOMA-IR distribution is used as a threshold to define children with high IR states. Here, mixed effect modeling is used to evaluate the association between IR and individual metabolites. The p-value was modeled in the R software (www.R-project. org) using the lmer function in software package lme4(Bates, Maechler et al, 2015) and calculated using Satterhwaite approximation implemented in the lmerTest software package (Kuznetsova, Brockhoff et al, 2016).

Example 2

Measurement of metabolite concentrations

Clinical and anthropometric characteristics of children at 5 and 14 years of age in the experimental study are summarized in table 1, and clinical and anthropometric characteristics of children at 5, 14 and 16 years of age in the main study are summarized in table 2. In both genders, HOMA-IR decreased until about 8 years of age, followed by an increase during puberty, a trend that was dependent on the schedule of peak height velocities (age. APHV interaction p < 0.001). IR was also positively correlated with BMI sds (p < 0.001).

Table 1: sex characteristics of the cohort 5 years old and 14 years old.

Data is median (quartile range)

Table 2: gender characteristics of the major cohort at 5, 14 and 16 years of age.

Data is median (quartile range)

Using data from all ages simultaneously, a mixed effect modeling was applied to evaluate the association between HOMA-IR and individual metabolites. In experimental studies, several metabolites including BCAAs, lipids and other amino acids showed significant association with HOMA-IR in longitudinal models (p <0.05), which is independent of BMI sds, physical activity and puberty schedules as shown in table 3. Table 4 reports the results of the same analysis performed on the main study cohorts.

Table 3: mixed effect model from examining the association between metabolites and insulin resistance in experimental studies And p value (n ═ 40)

Table 4: from a mixed effects model examining the association between metabolites and insulin resistance in the main study Estimate and p-value (n 150 ═ n)

The analysis highlights the importance of specific metabolites in amino acid, ketone body, glycolysis, and fatty acid metabolism in describing the changes in HOMA-IR throughout childhood. This is believed to be the first report of the metabolic contribution of a particular metabolic process to the overall change in insulin metabolism in a longitudinal and continuous manner.

Central energy-related metabolites

In the experimental and main study cohorts, mixed effects modeling described a reverse correlation of IR to the overall citrate and 3-D-hydroxybutyrate (p <0.001) and a positive correlation of IR to lactate (p < 0.01). The analysis of the main study described a statistically significant annual correlation of citrate (p <0.05 in the range of r 0.28 to r 0.66), whereas the statistically significant annual correlation of 3-D-hydroxybutyrate was not significant before 8 years of age and then significant (p <0.05 in the range of r 0.21 to r 0.58). In the main study, citrate was inversely correlated with IR at each cross-sectional time point between 5 and 16 years of age (correlation ranged from-0.21 to-0.52, p < 0.05). 3-D-hydroxybutyrate shows inverse cross-sectional relevance until age (relevance in the range of-0.21 to-0.53, p < 0.05). Lactate shows a positive cross-sectional correlation with IR (correlation in the range of r-0.13 to r-0.45, p < 0.05).

Metabolism of amino acids

Mixed effect modeling identified statistically significant inverse associations between histidine, creatine, and lysine and IR (p <0.05), which were repeated in the main study (p < 0.001). Each metabolite also showed inverse cross-sectional correlation with IR, particularly between 9 and 14 years of age (correlation in the range of-0.17 to-0.46 r, p < 0.05).

Lipid-related metabolites associated with IR

Of human serum1H-NMR spectroscopy allows monitoring of the signals associated with the fatty acyl groups bound to lipoproteins present in triglycerides, phospholipids and cholesterol esters, as well as the peaks from the glyceryl of triglycerides and the choline head group of phosphatidylcholine.

Here, the signal derived from methyl fatty acyl groups in choline-containing phospholipids shows a reverse correlation with IR, while the signal derived from methyl fatty acyl groups in LDL particles shows a positive correlation with IR. Lipid signals are highly correlated with each other (r >0.8 between 5 and 13 years of age, and r 0.6 at 14 years of age). These associations also exist for both the mixed effects model and the various time points in the main study. The cross-sectional associations between IR and phospholipids are inverse and statistically significant at age 7 (correlations in the range of-0.19 to-0.54), while those cross-sectional associations between IR and fatty acyl groups in LDL particles are positive and statistically significant between age 7 and age 14 (correlations in the range of 0.24 to 0.41). Although not significant in experimental studies (p 0.06), the fatty acyl groups in VLDL particles showed a positive association with IR in the mixed effect model, consistent with a cross-sectional association that was positive and statistically significant at 5 years of age and between 7 and 14 years of age (correlation in the range of r 0.25 to r 0.46).

Example 3

Metabolites indicative of higher HOMA-IR in adolescents

For each metabolite that showed a significant association with IR over time, the inventors further evaluated whether its serum concentration provided information on low IR status or high IR status in the 14-16 year old range. Optionally, the 91 th percentile of the HOMA-IR distribution is used as a threshold to define children with high IR states (Table 5). It was further explored which metabolites among those contributing most to the change in HOMA-IR in childhood could be an earlier and more indicative indicator of higher HOMA-IR in adolescence.

Table 5: estimated values and p-values from a mixed effects model examining the association between metabolites and the HOMA-IR set

Thus, among the most influential biochemicals that lead to high HOMA-IR in childhood, analysis indicates that:

● modeling of the mixing effect identifies the high IR state with lactate, fatty acyl groups in LDL and VLDL particles, and creatine: a significant positive correlation between glycine ratios over time.

● over time, a significant negative correlation was found between the high IR state and citrate, histidine, 3-D-hydroxybutyrate, glycine, creatine, lysine and phospholipids.

● fat mass (waist circumference) is also a statistically significant variable (p <0.001) in the high IR group that increases over time, with significant interaction between age and group (p < 0.001).

Mayer Davis et al have recently reported a significant increase in The annual Incidence of both Type 1and Type 2Diabetes in U.S. adolescents (10 to 19 years old) (inclusion Trends of Type 1and Type 2Diabetes mellitus events associated Youths, 2002-. It is well known that there are differences between ethnic and ethnic groups. As noted by Mayer Davis et al, this includes, by way of example, a relatively high increase in the incidence of type 2diabetes in ethnic and ethnic groups other than american non-hispanic whites. Variations across demographic subgroups may reflect different combinations of genetic, environmental and behavioral factors that contribute to diabetes. Accordingly, reference values should be generated for the proposed markers accordingly.

For example, in the study cohort, fold changes between groups were determined from population and provided at representative ages (table 6).

Table 6: fold change (percentage) in higher HOMA-IR individuals compared to the reference group

Age (age) 8.00 9.00 10.00 11.00 12.00 13.00 14.00 15.00 16.00
Lactate salt 11.68 4.89 11.50 8.52 13.14 13.50 12.01 18.69 13.19
Lysine 1.32 0.52 6.13 -2.77 -4.62 -0.02 -6.82 -3.61 2.47
Citric acid -4.66 -7.23 -5.50 -7.28 -6.13 -6.66 -16.67 -2.42 -1.07
Creatine -10.87 0.28 -3.78 -13.86 -6.72 -0.57 -10.57 -3.43 2.83
Glycine -3.83 -5.92 -7.94 -4.48 -10.02 -11.62 -15.75 -10.49 -6.74
Histidine 2.99 -6.44 -1.70 -3.22 -3.96 -9.30 -8.46 -3.54 -5.27

Overweight or obese people in children 5 years of age remain and progress further to excessive increases in fat mass and weight during adolescence and adolescence, and have higher HOMA-IR than other children. In particular, individuals in the 91 th percentile of juvenile HOMA-IR have particularly significantly lower histidine concentrations in serum over the age of 9, which corresponds to the time period during which the IR locus diverges between groups. They also exhibited higher body fat and central obesity (waist circumference) throughout childhood. Histidine status was inversely correlated with C-reactive protein levels at each age of the Earlybird population.

Our results also describe the remodeling of circulating phospholipid material throughout childhood, growth and development. This is a phenomenon well documented in the IR and T2D fields (Szymanska, Bouwman et al, 2012), but not excessive in childhood associated growth, development and fat gain. Remodeling of phospholipid material is often associated with a decrease in the concentration of ether-lipids (plasmablasts) associated with oxidative stress that has been reported in several diseases; such as diabetes, vascular disease and obesity.

Histidine and lysine are two representative targets for oxidative modification. Histidine is extremely sensitive to metal-catalyzed oxidation, producing 2-oxo-histidine and its ring cleavage products, while the oxidation of lysine produces carbonyl products such as aminoadipic semialdehyde. On the other hand, histidine and lysine are both nucleophilic amino acids and are therefore readily modified by electrophiles derived from lipid peroxidation (such as 2-enal, 4-hydroxy-2-enal and ketoaldehyde derived from lipid peroxidation). Histidine shows specific reactivity towards 2-enal and 4-hydroxy-2-enal, while lysine is a general target of aldehydes, leading to various types of adducts. Covalent binding of reactive aldehydes to histidine and lysine correlates with carbonyl reactivity and the appearance of protein antigenicity. None of these amino acids are reporters of IR in adult obese individuals.

Since inefficient lipolysis (high basal/low stimulation) is associated with future weight gain and impaired glucose metabolism and may constitute a therapeutic target (Arner, Andersson et al, 2018), our observations suggest unique nutritional requirements during growth and development to promote healthy fat metabolism. In the Earlybird cohort, overweight children at age 5 remain overweight throughout childhood, and will acquire a high IR status from age 10 during adolescent development and development of extra fat mass. Thus, our observations of negative associations of histidine, lysine and arginine status may indicate a potential imbalance in oxidative stress and adipocyte lipolysis during growth and development, which is accompanied or contributed to IR development.

Recommended dietary intake: 10 th edition (National Research Council.1989. recommended dietary intake: 10 th edition Washington, DC: The National academy of sciences Press).https://doi.org/10.17226/1349。)

● histidine is an essential amino acid in infants, but has not been recently shown to be required by adults (Cho et al, 1984; Kopple and Swendseid, 1981).

The requirement for histidine has not been quantified except during infancy. The requirement is difficult to determine, since the symptoms of the defect only occur after a long period of low intake. Kopple and Swendseid (1981) demonstrated that nitrogen balance decreased when histidine intake was less than 2 mg/kg/day and increased when intake increased to 4 mg/kg/day. WHO (1985) estimated the likely adult histidine requirement by extrapolation from infant requirements to be between 8 and 12mg/kg per day; this estimate may be high, but safe.

The requirement in mg/kg/day for adults is between 8 and 12.

From (Pencharz and Ball 2006),

■ the demand in mg/kg/day for children 2-10 years of age is about 12.

■ for children or adolescents between 10 and 14 years of age, the demand in mg/kg/day is about 12.

For adolescents between 14 and 18 years of age, the demand in mg/kg/day is about 11.

■ for adults, the demand in mg/kg/day is about 10.

● arginine is synthesized by mammals, but in insufficient quantities to meet the young requirements of most species. Although normal growth in human infants is believed to be undesirable, the need for arginine in preterm infants is unknown. Arginine synthesis may be insufficient to achieve adequate function of the urea cycle when arginine is present in small amounts relative to other amino acids, such as in intravenous solutions or amino acid mixtures, or when liver function is impaired (Heird et al, 1972).

● from (Pencharz and Ball 2006), the requirement for an alpha-amino acid consists of a number of components, namely the amount required for net incorporation into the protein plus the amount required for other biological processes. In particular, significant amounts of amino acids are required for the following, namely: (a) cysteine, glutamate and glycine for glutathione synthesis; (b) arginine for urea cycle activity, and (c) arginine, glycine and methionine for creatine synthesis.

● lysine is an essential amino acid in every stage of human life:

the demand in mg/kg/day for adults is about 12.

For children 10-12 years of age, the demand in mg/kg/day is about 44.

For a 2 year old child, the demand in mg/kg/day is about 64.

From (Pencharz and Ball 2006),

■ for children 2-10 years of age, the demand in mg/kg/day is about 35.

■ for children or adolescents between 10 and 14 years of age, the demand in mg/kg/day is about 35.

■ for a teenager of 14-18 years, the demand in mg/kg/day is about 33.

■ for adults, the demand in mg/kg/day is about 30.

Example 4

Juvenile amino acid and HOMA-IR status are related to adult HOMA-IR status

Using the spaerman correlation analysis for both insulin and HOMA-IR, the inventors described how insulin and HOMA-II states in children and adolescents were statistically significantly correlated with adult states throughout childhood and adolescence, from age 11 onwards (table 7). Therefore, the metabolites which have the greatest contribution to the change of the high HOMA-IR in childhood are more indicators of the higher HOMA-IR in childhood and are relevant markers of the high HOMA-IR state in adulthood.

Table 7: spearman correlation coefficient between a childhood individual parameter and the same individual parameter at age 20

Legend: spearman correlation analysis, data as r coefficient, p value, 95% CI, 99% CI, 99.9% CI

In addition, quantitative measurements of amino acids were made in serum samples from the same healthy individuals collected at 15 and 20 years of age to provide guidance on a healthy reference range.

Table 8: reference amino acid concentration in a reference group of subjects (N ═ 168)

Example 5

Juvenile amino acid status is associated with inflammatory status

Using the spearman correlation analysis, the inventors describe how the lysine, histidine, glycine and creatine to glycine ratios in teenagers are correlated with the marker C-reactive protein of inflammation (CRP, tables 9 and 10). Thus, the concentrations of metabolites associated with the HOMA-IR state in childhood are also associated with inflammatory states. Specifically, lysine, glycine, and histidine negatively correlated to HOMA-IR are also negatively correlated to CRP, while the creatine to glycine ratio is positively correlated to HOMA-IR and inflammatory states.

Table 9: correlation coefficient between metabolites and C-reactive protein in males

Table 10: correlation coefficient between metabolites and C-reactive protein in women

Age (age) 13.00 14.00
Lysine NS NS
Glycine NS NS
Creatine: ratio of Glycine 0.51(p=0.01) 0.39(p=0.21)
Histidine NS NS

Arner, p., d.p.andersson, j.backdahl, i.dahlman and m.ryden (2018). "Weight Gain and Impaired Glucose Metabolism in Women is Predicted by Inefficient Subcutaneous adipocyte Lipolysis (Weight Gain and affected Glucose Metabolism in Women area Predicted by ineffectiveness in surgery disease Cell Liposis). "Cell Metab

Bates, d., m.maechler, b.bolker, and s.walker (2015). "Linear Mixed Effect model (Fitting Linear Mixed-Effects Models Using lme4) was fitted Using lme 4. "Journal of Statistical Software 67(1):1-48.

Feng, r.n., y.c.niu, x.w.sun, q.li, c.zhao, c.wang, f.c.guo, c.h.sun and y.li (2013). "histidine supplements improve insulin resistance by inhibiting inflammation in obese women with metabolic syndrome: a random control experiment (inhibition of reactions in a reaction system with reaction of molecules in the reaction system) was performed. "Diabetologia 56(5):985-994.

Kuznetsova, a., p.b.brockhoff, and r.h.b.christensen (2016). "lmerTest: testing in the Linear Mixed Effect model (lmerTest: Tests in Linear Mixed Effects Models). And the version of the R software package is 2.0-30.https://CRAN.R-project.org/package=lmerTest.

Niu, y.c., r.n.feng, y.hou, k.li, z.kang, j.wang, c.h.sun, and y.li (2012). "Histidine and arginine are associated with inflammatory and oxidative stress in obese women (Histidine and arginine area associated with inflammatory and oxidative stress in obesity women). "Br J Nutr108(1):57-61。

Pencharz, p.b. and r.o.ball (2006). "Amino acid requirements of infants and children (Amino acid requirements of infants and childrens). "NestleNutr Workshop Ser Pediatr Program58: 109-116; discussion 116-.

Pinkney,J.、A.Streeter, j.hosking, m.mohammod, a.jeffery, and t.wilkin (2014). "childhood obesity, chronic inflammation and prepubertal decline of sex hormone-binding globulin: evidence related to the schedule of puberty (adaptability, respiratory inflammation, and the predictive decline of a set of hormone binding in childrens: evidence for associations with the timing of birth) (Earlybird 58). "J Clin Endocrinol Metab 99(9):3224-3232。

Szymanska, e., j.bouwman, k.strassburg, j.vervort, a.j.kangas, p.soinen, m.ala-Korpela, j.westerhuis, j.p.van Duynhoven, d.j.mela, i.a.macdonald, r.j.vresken, a.k.smile, and d.m.jacobs (2012). "gender-dependent association of metabolite profile and body fat distribution in healthy population with central obesity: towards metabolomics diagnostics (population-dependent associations of metabolic profiles and body fat distribution in a help distribution with central biology: towards metabolomics diagnostics). "OMICS 16(12):652-667。

26页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:包含罗丹宁衍生物作为活性成分的用于预防、减轻或治疗骨关节炎的药物组合物

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!