WO2024178621A1 - Use of compounds in preventing and/or treating obesity or nafld - Google Patents
Use of compounds in preventing and/or treating obesity or nafld Download PDFInfo
- Publication number
- WO2024178621A1 WO2024178621A1 PCT/CN2023/078793 CN2023078793W WO2024178621A1 WO 2024178621 A1 WO2024178621 A1 WO 2024178621A1 CN 2023078793 W CN2023078793 W CN 2023078793W WO 2024178621 A1 WO2024178621 A1 WO 2024178621A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- drug
- subject
- compound
- 4hpaa
- nafld
- 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
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/185—Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
- A61K31/19—Carboxylic acids, e.g. valproic acid
- A61K31/192—Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/04—Anorexiants; Antiobesity agents
Definitions
- the present invention pertains to the field of medical technology.
- the present invention relates to use of a compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in in preparation of a drug or a dietary supplement.
- the drug is used for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in a subject, and the dietary supplement is used for weight management or weight control of a subject.
- NAFLD nonalcoholic fatty liver disease
- Excess body fat is one of the greatest public health problems in this era. According to the World Health Organization, global obesity has tripled since 1975. In 2016, over 1.9 billion adults and 340 million children or adolescents aged 5-19 years in the world were overweight or obese. Obesity would further promote the risk of various serious diseases, including but not limited to cardiovascular disorders, type 2 diabetes, musculoskeletal conditions, and certain cancers.
- Obesity is a complex disease that results from the interplay of genetic, behavioral, and environmental factors. Recent studies show that the gut microbiota is a pivotal environmental factor influencing body weight gain in both rodent and human population studies, exerting either protective or detrimental effects on the host. A great number of cohort studies focusing on the gut microbiota with obesity have been accomplished, but controversy regarding the relative abundance of bacteria and obesity remains. Certain bacterial species have been shown to exhibit inhibitory effects on obesity, such as Akkermansia muciniphila and Parabacteroides distasonis, while many others promote obesity. The exact mechanisms for microbiota to affect body fat accumulation could be complicated.
- AAA aromatic amino acids
- Trp tryptophan
- Phe phenylalanine
- Tyr tyrosine
- the immunohistological analysis showed increased Myd88 in intestinal immune cells from 4HPAA-treated mice, further supporting the notion that AAA metabolites prevent obesity by modulating intestinal immune control.
- the inventions as follows are provided.
- the present application provides use of a compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in preparation of a drug, wherein the drug is used for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in a subject; the drug is administrated to the subject by gastrointestinal or parenteral administration;
- a compound of formula (I) pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in preparation of a drug, wherein the drug is used for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in a subject; the drug is administrated to the subject by gastrointestinal or parenteral administration;
- n is an integer selected from 1-6 (e.g. 1, 2, 3, 4, 5 or 6) ;
- the compound isn’ t 4-hydroxyphenylacetic acid (4HPAA) in condition that the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof is parenterally administrated to the subject for preventing and/or treating NAFLD.
- 4HPAA 4-hydroxyphenylacetic acid
- n in formula (I) is 1 or 2.
- - (CH2) n-COOH in formula (I) is at the para-or meta-position of phenol hydroxyl.
- the compound is selected from 4-hydroxyphenylpyruvic acid (4HPPA) , 3-hydroxyphenylpropionic acid (3HPP) and 4-hydroxyphenylpropionic acid (4HPP) of which the chemical structures are shown below,
- the drug can be administered by any suitable method known in the art, including gastrointestinal or parenteral administration.
- the gastrointestinal administration is oral administration or nasal feeding.
- the drug may be in any dosage form suitable for gastrointestinal or parenteral administration.
- the drug is in a liquid dosage form (e.g. emulsion, microemulsion, solution, suspension, syrup or elixir) or a solid dosage form (e.g. capsule, tablet, pill, lozenge, powder or granule) for oral administration.
- a liquid dosage form e.g. emulsion, microemulsion, solution, suspension, syrup or elixir
- a solid dosage form e.g. capsule, tablet, pill, lozenge, powder or granule
- the preferred dosage form depends on the intended mode of administration and therapeutic use.
- Liquid dosage form for oral administration includes pharmaceutically acceptable emulsion, microemulsion, solution, suspension, syrup, elixir and the like.
- the liquid dosage form may contain an inert diluent commonly used in the art, such as water or other solvent, solubilizer and emulsifier, such as ethanol, isopropanol, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butanediol, dimethylformamide, oil (e.g., cottonseed oil, peanut oil, corn oil, germ oil, olive oil, castor oil and sesame oil) , glycerin, tetrahydrofurfuryl alcohol, polyethylene glycol, and fatty acid ester of sorbitan as well as mixtures thereof.
- solubilizer and emulsifier such as ethanol, isopropanol, ethyl acetate, benzyl alcohol, benzyl be
- liquid dosage form for oral administration may also include adjuvant, such as wetting agent, emulsifier and suspending agent, sweetening agent, flavoring agent, and fragrance.
- adjuvant such as wetting agent, emulsifier and suspending agent, sweetening agent, flavoring agent, and fragrance.
- Solid dosage form for oral administration includes capsule, tablet, pill, lozenge, powder, granule and the like.
- the solid dosage form may contain pharmaceutically acceptable inert excipient or carrier, such as filler (e.g., lactose, sucrose, glucose, mannitol, starch, microcrystalline cellulose, galactose, crospovidone and calcium sulfate) ; binder (e.g., carboxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and acacia) ; wetting agent (e.g., cetyl alcohol and glyceryl monostearate) ; disintegrant (e.g., agar, calcium carbonate, starch, alginic acid, sodium carboxymethyl cellulose, sodium carboxymethyl starch) ; lubricant (e.g., talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium laurel sulfate) ; and mixtures thereof.
- filler e.g., lactose, sucrose, glucose, manni
- the compound or pharmaceutical composition of the present invention can also be administered by a non-oral route.
- parenteral administration for example, subcutaneous injection, intravenous injection, intraperitoneal injection, intramuscular injection, intrasternal injection, and infusion.
- the dosage form for parenteral administration may be an injection preparation, including injection solution, sterile powder for injection, or concentrated solution for injection.
- the injection dosage form may contain a pharmaceutically acceptable carrier such as sterile water, Ringer's solution and isotonic sodium chloride solution.
- An appropriate additive such as antioxidant, buffer and bacteriostatic agent may also be added depending on the nature of the drug.
- the pharmaceutical composition of the present invention can be prepared by any well-known pharmaceutical process, such as effective formulation and administration method.
- effective formulation and administration method are well known in the art and described in standard textbooks.
- the formulation of pharmaceuticals is described in, for example, Hoover, John E., Remington's Pharmaceutical Sciences. Mack Publishing Co., Easton, Pennsylvania, 1975; Liberman et al., Pharmaceutical Dosage Forms, Marcel Decker, New York, NY, 1980; and Kibbe et al., Handbook of Pharmaceutical Excipients (3rd edition) , American Pharmaceutical Association, Washington, 1999.
- the drug is used for preventing and/or treating obesity, overweight, and the drug is administrated to the subject by gastrointestinal administration (e.g. oral administration) .
- gastrointestinal administration e.g. oral administration
- the drug is used for preventing and/or treating nonalcoholic fatty liver disease (NAFLD) in the subject; the drug is administrated to the subject by parenteral administration; the n in formula (I) is an integer selected from 2-6.
- NAFLD nonalcoholic fatty liver disease
- n in formula (I) is 2 or 3.
- the compound is 3HPP or 4HPP.
- the compound of formula (I) is found to have protective effect on the liver. Particularly, it was found that IP-injected the compound (e.g. 3HPP) protected a subject from HFD-induced hepatic steatosis. Therefore, the compound of formula (I) may serve as a drug candidate for the prevention or therapeutics of nonalcoholic fatty liver disease (NAFLD) .
- NAFLD nonalcoholic fatty liver disease
- NAFLD includes simple fatty liver and non-alcoholic steatohepatitis (NASH) .
- the compound of formula (I) may also be used in preparation of a dietary supplement, wherein the dietary supplement is used for weight management or weight control of a subject.
- the dietary supplement may be in a liquid dosage form (e.g. emulsion, microemulsion, solution, suspension, syrup or elixir) or a solid dosage form (e.g. capsule, tablet, pill, lozenge, powder or granule) for oral administration as described above.
- the present application provides use of the compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in preparation of a drug or dietary supplement, wherein the drug or dietary supplement is used for modulation of intestinal immune control in a subject.
- the modulation of intestinal immune control in a subject may comprises (1) inducing IgA production in the subjetct’s intestine, (2) suppressing expression of a key factor (e.g. fatty acid binding protein 2 (Fabp2) , fatty acid binding protein 4 (Fabp4) , stearoyl-CoA desaturase 1 (Scd1) , lipoprotein lipase (Lpl) and/or cluster of differentiation 36 (Cd36) ) for intestinal lipid absorption in the subject’s intestine, (3) suppressing inflammation in the subjetct’s intestine, (4) increasing MyD88 expression in the subject’s intestinal immune cells, and/or (5) increasing number of immune cells (e.g. B cells and/or Th17 cells) in the subjetct’s intestinal mucosa.
- a key factor e.g. fatty acid binding protein 2 (Fabp2) , fatty acid binding protein 4 (Fabp4) , stearoyl-CoA desaturase 1 (Scd
- the drug or dietary supplement is used for suppressing lipid absorption in the subject.
- the drug is used for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in the subject.
- NASH nonalcoholic fatty liver disease
- the dietary supplement is used for weight management or weight control of a subject.
- the compound of formula (I) may be used in combination with other pharmaceutically active agents. Therefore, the drug described above may further comprises an additional pharmaceutically active agent, e.g. anti-diabetic drug, anti-obesity drug, anti-hypertensive drug, anti-atherosclerosis drug, lipid-lowering drug or anti-inflammatory drug.
- an additional pharmaceutically active agent e.g. anti-diabetic drug, anti-obesity drug, anti-hypertensive drug, anti-atherosclerosis drug, lipid-lowering drug or anti-inflammatory drug.
- the compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof and the additional pharmaceutically active agent are provided as separate components or mixed components. Therefore, compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof and the additional pharmaceutically active agent can be administered simultaneously, separately or sequentially.
- the drug described above may comprise a "therapeutically effective amount” or “prophylactically effective amount” of compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof.
- a “prophylactically effective amount” refers to an amount sufficient to prevent, stop, or delay the occurrence of a disease.
- therapeutically effective amount refers to an amount sufficient to cure or at least partially prevent a disease and complication thereof in a patient who has already suffered from the disease.
- compound of formula (I) may have a therapeutically effective amount varying according to the following factors: severity of disease to be treated, overall state of patient’s own immune system, patient’s general conditions such as age, weight and gender, administration mode of drug, and other treatments simultaneously administered, etc.
- the drug or dietary supplement comprises the compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in an amount of 0.01 mg to 10 g, e.g. 0.01 mg to 0.1 mg, 0.1 mg to 1 mg, 1 mg to 10 mg, 10 mg to 100 mg, 100 mg to 1000 mg, 1 g to 3 g, 3 g to 5 g, 5 g to 7 g, or 7 g to 10 g.
- the present application provides a method for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in a subject, or controlling or managing weight in a subject, wherein the method comprises administrating a compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof by gastrointestinal administration (e.g. oral administration or nasal feeding) or parenteral administration to the subject; the compound isn’ t 4-hydroxyphenylacetic acid (4HPAA) in condition that the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof is parenterally administrated to the subject for preventing and/or treating NAFLD.
- gastrointestinal administration e.g. oral administration or nasal feeding
- 4HPAA 4-hydroxyphenylacetic acid
- the method is for preventing and/or treating obesity or overweight in the subject; the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal is administrated to the subject by gastrointestinal administration (e.g. oral administration) .
- gastrointestinal administration e.g. oral administration
- the method is for preventing and/or treating nonalcoholic fatty liver disease (NAFLD) in the subject; the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal is administrated to the subject by parenteral administration; the n in formula (I) is an integer selected from 2-6.
- NAFLD nonalcoholic fatty liver disease
- the method is for controlling or managing weight in the subject; the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof is administrated to the subject by gastrointestinal administration (e.g. oral administration) .
- gastrointestinal administration e.g. oral administration
- the present application provides a method for modulation of intestinal immune control in a subject, wherein the method comprises administrating a compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof by gastrointestinal administration (e.g. oral administration or nasal feeding) or parenteral administration to the subject.
- gastrointestinal administration e.g. oral administration or nasal feeding
- parenteral administration e.g. parenteral administration to the subject.
- the modulation of intestinal immune control in a subject comprises (1) inducing IgA production in the subjetct’s intestine, (2) suppressing expression of a key factor (e.g. Fabp2, Fabp4, Scd1, Lpl and/or Cd36) for intestinal lipid absorption in the subject’s intestine, (3) suppressing inflammation in the subjetct’s intestine, (4) increasing MyD88 expression in the subject’s intestinal immune cells, and/or (5) increasing number of immune cells (e.g. B cells and/or Th17 cells) in the subjetct’s intestinal mucosa.
- a key factor e.g. Fabp2, Fabp4, Scd1, Lpl and/or Cd36
- the method is for suppressing lipid absorption in the subject.
- the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof may be administrated in a liquid dosage form (e.g. emulsion, microemulsion, solution, suspension, syrup or elixir) or a solid dosage form (e.g. capsule, tablet, pill, lozenge, powder or granule) .
- a liquid dosage form e.g. emulsion, microemulsion, solution, suspension, syrup or elixir
- a solid dosage form e.g. capsule, tablet, pill, lozenge, powder or granule
- Any one of the methods described above may also comprise: administering the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in combination with an additional therapy.
- the additional therapy can be any therapy known for metabolic diseases, such as surgery, targeted therapy, immunotherapy, hormone therapy or gene therapy. This additional therapy can be administered before, simultaneously or after the administration of compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof as described herein.
- the additional therapy comprises administration of an additional pharmaceutically active agent selected from the group consisting of anti-diabetic drug, anti-obesity drug, anti-hypertensive drug, anti-atherosclerosis drug, lipid-lowering drug and anti-inflammatory drug.
- an additional pharmaceutically active agent selected from the group consisting of anti-diabetic drug, anti-obesity drug, anti-hypertensive drug, anti-atherosclerosis drug, lipid-lowering drug and anti-inflammatory drug.
- the method is used in combination with a reduced-calorie diet and/or increased physical activity.
- the dosage regimen can be adjusted to obtain the best objective response (e.g., therapeutic or preventive response) .
- it can be administered in a single dose, can be administered multiple times over a period of time, or the dose can be reduced or increased proportionally to the urgency of the treatment situation.
- the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof is administrated to the subject once, twice or three times a day.
- the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof has a therapeutically or prophylactically effective amount with a typical and non-limiting range of 0.01 to 500 mg/kg/day, e.g. 0.01 to 1 mg/kg/day, 1 to 20 mg/kg/day, 20 to 50 mg/kg/day, 50 to 100 mg/kg/day, 100 to 200 mg/kg/day, or 200 to 500 mg/kg/day. It should be noted that the dosage may vary with the type and severity of the symptoms to be treated.
- the term "pharmaceutically acceptable salt” refers to (i) a salt formed by an acidic functional group (e.g., -COOH) present in the compound provided by the present invention and an appropriate inorganic or organic cation (base) , which includes, but is not limited to, alkali metal salt, such as sodium salt, potassium salt, lithium salt, etc.; alkaline earth metal salt, such as calcium salt, magnesium salt, etc.; other metal salt, such as aluminum salt, iron salt, zinc salt, copper salt, nickel salt, cobalt salt, etc.; inorganic alkali salt, such as ammonium salt; organic alkali salt, such as tert-octylamine salt, dibenzylamine salt, morpholine salt, glucosamine salt, phenylglycine alkyl ester salt, ethylenediamine salt, N-methylglucamine salt, guanidine salt, diethylamine salt, triethylamine salt, dicyclohexyl
- ester refers to an ester formed by -COOH present in the compound provided by the present invention and an appropriate alcohol, or an ester formed by -OH present in the compound provided by the present invention and an appropriate acid (e.g., a carboxylic acid or an oxygen-containing inorganic acid) .
- Suitable ester groups include, but are not limited to, formate, acetate, propionate, butyrate, acrylate, ethylsuccinate, stearyl fatty acid ester, or palmitate. In the presence of an acid or base, the ester can undergo a hydrolysis reaction to generate the corresponding acid or alcohol.
- solvate refers to a substance formed by the association of the compound of the present invention with a solvent molecule.
- the solvent may be an organic solvent (e.g., methanol, ethanol, propanol, acetonitrile, etc. ) , for example, the compound of the present invention may form an ethanolate with ethanol.
- the compound of the present invention can also form an hydrate with water.
- crystal form refers to a crystal structure of a substance.
- the compound of the present invention may exist in one crystal structure or in multiple crystal structures, that is, have a "polymorphic form” .
- the compound of the present invention may exist in different crystal forms.
- stereoisomer includes conformational isomer and configurational isomer, wherein the configurational isomer mainly includes cis-trans isomers and optical isomers.
- the compound of the present invention may exist in the form of stereoisomer, and therefore encompass all possible stereoisomeric forms, and any combination or any mixture thereof, for example, a single enantiomer, a single diastereomer or a mixture of the above.
- the compound of the present invention comprises an alkene double bond, unless otherwise specified, it includes cis isomer and trans isomer, and any combination thereof.
- the term "pharmaceutically acceptable carrier or excipient” refers to a carrier and/or excipient that is pharmacologically and/or physiologically compatible with the subject and the active ingredient, which is well known in the art (see, for example, Remington's Pharmaceutical Sciences. Edited by Gennaro AR, 19th ed. Pennsylvania: Mack Publishing Company, 1995) , and includes, but is not limited to: disintegrant, binder, surfactant, glidant, lubricant, pH adjuster, ionic strength enhancer, agent for maintaining osmotic pressure, agent for delaying absorption, diluent, antioxidant, coloring agent, flavoring agent, preservative, taste masking agent, etc.
- non-limiting examples of disintegrant include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methylcellulose, microcrystalline cellulose, lower alkyl-substituted hydroxypropylcellulose, starch, pregelatinized starch and sodium alginate.
- Non-limiting examples of binder include microcrystalline cellulose, gelatin, sugar, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinized starch, hydroxypropyl cellulose, and hydroxypropyl methyl cellulose.
- Non-limiting examples of diluent include lactose (monohydrate, spray-dried monohydrate, anhydrous, etc. ) , mannitol, xylitol, glucose, sucrose, sorbitol, microcrystalline cellulose, starch, and calcium hydrogen phosphorate dihydrate.
- Non-limiting examples of surfactant include sodium lauryl sulfate and polysorbate 80.
- Non-limiting examples of glidant include silica and talc.
- Non-limiting examples of lubricant include magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate and sodium lauryl sulfate.
- Non-limiting example of pH adjusting agent includes but is not limited to phosphate buffer.
- Ionic strength enhancer includes, but is not limited to, sodium chloride.
- Agent for maintaining osmotic pressure includes but is not limited to sugar, NaCl and the like.
- Agent that delays absorption includes, but is not limited to, monostearate and gelatin.
- Preservative includes, but is not limited to, various antibacterial agents and antifungal agents, such as thimerosal, 2-phenoxyethanol, paraben, chlorobutanol, phenol, sorbic acid and the like.
- prevention refers to a method performed to prevent or delay the occurrence of a disease or disorder or symptom in a subject.
- treatment refers to a method performed to obtain a beneficial or desired clinical result.
- the beneficial or desired clinical result includes, but is not limited to, alleviating symptom, narrowing disease scope, stabilizing (i.e., not getting worse) disease state, delaying or slowing disease development, improving or alleviating disease state, and relieving symptom (whether partially or fully) , whether detectable or undetectable.
- treatment may also refer to that a survival period is prolonged compared to an expected survival period (if not receiving treatment) .
- the term "subject" refers to a mammal, such as a primate mammal, such as a human.
- FIG. 1 Oral intake of 4HPAA, 3HPP, and 4HPP suppress weight gain in HFD-fed mice.
- A Schematic design of oral treatment in the HFD mouse model.
- B A presentative image of ND-fed, HFD-fed, and HFD-fed plus 4HPAA-treated mice.
- C The weight gain of ND-fed and HFD-fed mice treated with or without 4HPAA 8 weeks was measured and plotted on the chart.
- D The body fat percentage of ND-fed and HFD-fed mice treated with or without 4HPAA for 8 weeks was measured and plotted on the chart.
- E The food intake of HFD-fed mice treated with or without 4HPAA after 8 weeks was measured and plotted on the chart.
- FIG. 1 Metabolic performances of the HFD-fed mice orally treated with or without 4HPAA.
- A Daily food intake of these mice during the treatment.
- FIG. 3 4HPAA intervention blocks the body weight gain in mice already become obese.
- A Schematic view of the experiment design. Two groups of mice were fed with HFD under the same condition for 14 weeks. After 8 weeks, one group was kept given normal water (control) and the other was changed to 4HPAA-containing water for the next 6 weeks. The mouse's body weight was measured weekly.
- FIG. 4 Similar fecal metabolome was observed in 4HPAA, 4HPP, and 3HPP treatments.
- A The Venn diagram shows that the upregulated metabolites in the 4HPAA, 4HPP, and 3HPP groups are largely overlapped.
- B The heat map shows the overlapped upregulated metabolites in (A) .
- Figure 5 4HPAA, 3HPP, and 4HPP alleviate adipocyte hypertrophy and hepatic steatosis.
- A Representative images of H&E stained adipose tissues, including iBAT, iWAT, and eWAT, from ND/HFD-fed mice orally treated with or without 4HPAA, 4HPP, 3HPP, and tyrosol after 10 weeks.
- B-D The average diameters of adipocytes in iBAT (B) , iWAT (C) , and eWAT (D) were measured and plotted on the charts.
- E Representative images of H&E stained liver sections from ND/HFD-fed mice orally treated with or without 4HPAA, 4HPP, 3HPP, and tyrosol after 10 weeks.
- F The fatty liver histological scores in (E) were assessed and plotted on the diagram.
- FIG. 7 IP injected 4HPAA and 3HPP failed to inhibit body fat accumulation in HFD-fed mice.
- A Schematic design of IP treatment of 4HPAA/3HPP in the HFD mouse model.
- B-C ND/HFD-fed mice were IP injected with or without 4HPAA and 3HPP twice a week. After 12 weeks, the body weight gain (B) and fat percentage (C) of these mice were assessed and plotted on the charts.
- D Representative images of H&E stained adipose tissues, including iBAT, iWAT, and eWAT, from ND/HFD-fed mice IP injected with saline, 4HPAA, and 3HPP for 12 weeks.
- Figure 8 4HPAA blocks lipid absorption and induces IgA production in the gut.
- A Volcano plot of colon transcripts. 63 upregulated gene hits and 173 downregulated gene hits (with foldchange over 2 and p-value less than 0.05) were highlighted in red and blue respectively.
- B Gene set enrichment analysis based on the upregulated and downregulated gene hits.
- C The heat map displays 25 top-upregulated genes, which all encode immunoglobulins, in the 4HPAA-treated HFD-fed mice compared with control HFD-fed mice.
- D The heat map displays all downregulated genes. Notable genes related to lipid absorption and metabolism are marked with blue labels.
- RT-PCR analysis shows that the transcription of Cd36 and Scd1 was drastically inhibited in the 4HPAA-fed mice compared to control mice.
- IHC analysis shows the reduced protein level of Cd36 in the 4HPAA-fed mice compared to control mice.
- G Immunofluorescence analysis shows increased mucosa infiltration of B cells (anti-IgA, upper panel) and Th17 cells (anti-IL17, lower panel) in HFD-fed mice orally treated with 4HPAA or 3HPP.
- IHC analysis shows increased Myd88 expression in immune cells (marked by red arrows) of HFD-fed mice orally treated with 4HPAA.
- Figure 10 Schematic drawing of the proposed model for immune control by microbial AAA metabolites in the intestinal epithelium.
- Mouse feed, reagents, and antibodies were purchased from the commercial vendors: High Fat Diet (OpenSource Diets, #D12492) , Normal Diet (OpenSource Diets, #D12450J) , Brain Heart Infusion (Solarbio, Cat#B8130) , 4HPAA (Sigma-Aldrich, Cat#H50004) , 3HPP (Leyan, Cat#1062804) , 4HPP (Leyan, Cat#1033824) , tyrosol (Cayman, Cat#27600) , acetonitrile (J&K, Cat#75-05-8) , Antifade Mounting Medium with DAPI (Beyotime, Cat#P0131) , rabbit monoclonal antibodies against MyD88 (GeneTex, Cat#GTX112987) and Cd36 (ABclonal, Cat#A19016) , FITC labeled rabbit monoclonal antibody against IgA (Biolegend, Cat#1165-02) , Fluor
- SPF pathogen free mice
- mice Male, aged 6-8 weeks
- All mice housed at 20–24 °C with 40–60%humidity and had a 12 h cycle of light/darkness (7 a.m. to 7 p.m. ) .
- All animal protocols were approved by the Institutional Animal Care and Use Committee (IACUC) of Westlake University.
- mice were fed a HFD (60%fat) or ND (10%fat) .
- HFD 50%fat
- ND 50%fat
- 4HPAA 3HPP
- 4HPP 4HPP
- tyrosol tyrosol
- IP injection the dosage of 4HPAA or 3HPP was a series of gradient diluted concentrations: 8, 2.8, 0.9 and 0.3 mg/kg, each of which were intraperitoneal injected twice a week for 8 weeks.
- mouse body weight was measured at the beginning and weekly afterwards. At specific time points, fresh fecal samples were collected and stored at -80°C.
- Fat mass values were assessed at the end of each experiment using EchoMRI 100H Body Composition Analyzer (EchoMRI, USA) . Serum sample of each mouse was collected at the terminal experiment and freshly detected (HITACHI Automatic Aralyzer, Cat#3100) for the biochemical items mentioned in this study, including AST (aspartate transaminase) , ALT (alanine transaminase) , LDL-C (low-density lipoprotein cholesterol) , HDL-C (high-density lipoprotein cholesterol) , and TC (total cholesterol) .
- AST aspartate transaminase
- ALT alanine transaminase
- LDL-C low-density lipoprotein cholesterol
- HDL-C high-density lipoprotein cholesterol
- TC total cholesterol
- mice were sacrificed and their liver, colon, and adipose tissues were fixed in formalin for 12 hours, dehydrated with gradient alcohol, cleared with xylene, and embedded in paraffin. Paraffin blocks were cut into 5 ⁇ m thick sections and either stained with H&E or detected with specific antibodies.
- Cd36 and MyD88 (anti-Cd36: 1: 200; anti-MyD88: 1: 500) were detected by immunochemistry analysis and IgA-positive B cells and IL-17-positive T cells (anti-IgA: 1: 400; anti-IL-17: 1: 400) were detected by immunofluorescence analysis.
- the images were captured using either an upright Nikon Motorized Fluorescence Microscope (for H&E and IHC) or Nikon Spinning-disk Confocal Microscope (for IF) .
- the pathology was assessed and scored by two pathologists blinded on a scale of 0 to 4 (normal, minimal, mild, moderate, and severe) .
- fecal DNA samples were extracted using the soil DNA kit (Omega Bio-Tek, Norcross, GA, U.S. ) , according to manufacturer instructions.
- the upstream primer 338F (5 '-actCCTACGGgaggCAGCAGCAG-3') carrying barcode sequence and downstream primer 806R (5 '-GGACTACHVGGGTWTCTAAT-3') were used to target the V3-V4 variable region of 16S rRNA gene.
- the NEXTFLEX Rapid DNA-Seq Kit was used to construct the library of the purified PCR products, and sequencing was performed using Illumina's Miseq PE300/NovaSeq PE250 platform (Shanghai Majorbio Bio-pharm Biotechnology Co., Ltd. ) .
- Fastq-files were demultiplexed by the MiSeq Controller Software (Bcl2fastq version 2.20, Illumina Inc. ) .
- the sequences were trimmed for amplification primers, and sequencing adapters, merge-paired, and quality filtered by Quantitative Insights into Microbial Ecology (QIIME) software version 2-2020.2.
- DADA2 was used for amplicon sequence variants (ASVs) clustering equaling 100%.
- a representative sequence was picked for each ASV and the Sliva reference database version 138 was used to annotate taxonomic information.
- the absolute abundance table was extracted from the pipeline and converted to relative abundances by normalizing for analyzing the composition of gut microbiota by QIIME2.
- mice were sacrificed after 3 months of the oral treatment and freshly collected colon tissue sections (50-100 mg) were quick-frozen and stored in liquid nitrogen. Following RNA purification, reverse transcription, library construction, and sequencing were performed at Shanghai Majorbio Bio-pharm Biotechnology Co., Ltd. (Shanghai, China) .
- Differential expression analysis based on the expression of quantitative results, analysis of genetic variations between groups, gain occurred between the two groups of differentially expressed genes, variance analysis software for DESeq2 (3.16) , the threshold value is:
- > 1 &p-value ⁇ 0.05.
- mice Colon tissues from sacrificed mice after 3 months of the oral drug trial were dissected to 50-100mg and were slightly washed with 0.9%saline.
- Total RNA isolation of mice colon tissues has followed the manuscript of a MolPure Cell/Tissue Total RNA Kit (Yeasen, China) .
- Reverse transcription was performed using an ABScript III RT Master Mix for qPCR with gDNA Remover (ABclonal, Cat#RK20429) from 2 ⁇ g of total RNA.
- mice were fed with either a normal diet (ND) or HFD and routinely given water (control) or water containing 1.5 mg/ml ( ⁇ 10 mM) 4HPAA for 8 weeks (Figure 1A) .
- the average body weight of 4HPAA-treated HFD-fed mice was drastically lower than the non-treated HFD-fed mice and similar to the ND-fed mice ( Figure 1B) .
- 4HPAA-treated HFD-fed mice ( ⁇ 7.90 g) were close to the ND-fed mice either treated ( ⁇ 6.40 g) or non-treated ( ⁇ 6.64 g) with 4HPAA, but is ⁇ 45%less than the non-treated HFD-fed mice ( ⁇ 14.31 g) ( Figure 1C) .
- Interscapular brown adipose tissue iBAT
- inguinal white adipose tissue iWAT
- epididymal white adipose tissue eWAT
- adipocytes in the iBAT, iWAT, and eWAT from the HFD-fed mice were larger compared to the ones from the ND-fed mice ( Figures 5A-D) .
- Such adipocyte hypertrophy is a typical phenotype of obesity.
- adipocytes from the HFD-fed mice treated with 4HPAA, 3HPP, and 4HPP showed smaller sizes, which is more similar to the ND-fed mice.
- Oral feeding of tyrosol failed to rescue adipocyte hypertrophy in the HFD-fed mice ( Figures 5A-D) .
- adipocytes in the iBAT from 4HPAA, 3HPP, or 4HPP-treated HFD-fed mice were even slightly smaller than the ones from the ND-fed mice.
- the brown adipocytes might be more sensitive upon 4HPAA, 3HPP, or 4HPP treatment.
- HFD-induced obesity is normally accompanied by the nonalcoholic fatty liver, also called hepatic steatosis.
- 4HPAA, 3HPP, and 4HPP feeding all effectively alleviated the hepatic steatosis induced by HFD, while tyrosol had no protective effects ( Figures 5E-F) .
- Classic serum biomarkers for liver injury and blood lipid disorder were further measured, including ALT, AST, LDL, HDL, and TC.
- the 4HPAA, 3HPP, or 4HPP-treated HFD-fed mice had lower TC and LDL compared to the control HFD-fed. No obvious difference was observed for ALT, AST, and HDL ( Figure 6) .
- RNA-seq RNA-sequencing
- Figure 8A and Figure 9 RNA-sequencing
- GSEA Gene set enrichment analysis
Landscapes
- Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Child & Adolescent Psychology (AREA)
- Obesity (AREA)
- Hematology (AREA)
- Diabetes (AREA)
- Gastroenterology & Hepatology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicinal Preparation (AREA)
Abstract
The present invention pertains to the field of medical technology. In particular, the present invention relates to use of compounds in prevent and/or treat obesity, overweight or non-alcoholic fatty liver disease (NAFLD).
Description
The present invention pertains to the field of medical technology. In particular, the present invention relates to use of a compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in in preparation of a drug or a dietary supplement. The drug is used for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in a subject, and the dietary supplement is used for weight management or weight control of a subject.
Excess body fat is one of the greatest public health problems in this era. According to the World Health Organization, global obesity has tripled since 1975. In 2016, over 1.9 billion adults and 340 million children or adolescents aged 5-19 years in the world were overweight or obese. Obesity would further promote the risk of various serious diseases, including but not limited to cardiovascular disorders, type 2 diabetes, musculoskeletal conditions, and certain cancers.
Obesity is a complex disease that results from the interplay of genetic, behavioral, and environmental factors. Recent studies show that the gut microbiota is a pivotal environmental factor influencing body weight gain in both rodent and human population studies, exerting either protective or detrimental effects on the host. A great number of cohort studies focusing on the gut microbiota with obesity have been accomplished, but controversy regarding the relative abundance of bacteria and obesity remains. Certain bacterial species have been shown to exhibit inhibitory effects on obesity, such as Akkermansia muciniphila and Parabacteroides distasonis, while many others promote obesity. The exact mechanisms for microbiota to affect body fat accumulation could be complicated.
Contents of the Present Invention
Proteolysis of food in the gastrointestinal tract generates large amounts of aromatic amino acids (AAA) , including tryptophan (Trp) , phenylalanine (Phe) , and tyrosine (Tyr) , which provides a rich source for gut microbes to generate a vast number of aromatic compounds. One common gut-microbial metabolic pathway for Phe/Tyr is AAA aminotransferase-mediated
transamination, where Tyr is first metabolized to 4-hydroxyphenylpyruvic acid (4HPPA) and later turned to 4-hydroxyphenylacetic acid (4HPAA) , 4-hydroxyphenyllactic acid (4HPLA) , 4-hydroxyphenylpropionic acid (4HPP) , 4-methylphenol (p-cresol) , and/or 4-hydroxyphenylethanol (tyrosol) .
The inventors found that the microbial AAA metabolism pathways appeared to be associated with body fat accumulation. Three AAA metabolites, 4HPAA, 4HPP, and 3HPP, effectively protected mice from a high-fat diet (HFD) -induced obesity. Full protection against obesity was achieved preferably via oral delivery of 4HPAA and 3HPP. Transcriptomics analysis revealed that 4HPAA induced IgA production and suppressed lipid uptake and inflammation in the intestines. Moreover, the immunohistological analysis showed increased Myd88 in intestinal immune cells from 4HPAA-treated mice, further supporting the notion that AAA metabolites prevent obesity by modulating intestinal immune control. The inventions as follows are provided.
In one aspect, the present application provides use of a compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in preparation of a drug, wherein the drug is used for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in a subject; the drug is administrated to the subject by gastrointestinal or parenteral administration;
wherein n is an integer selected from 1-6 (e.g. 1, 2, 3, 4, 5 or 6) ;
the compound isn’ t 4-hydroxyphenylacetic acid (4HPAA) in condition that the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof is parenterally administrated to the subject for preventing and/or treating NAFLD.
In some embodiments, the n in formula (I) is 1 or 2.
In some embodiments, - (CH2) n-COOH in formula (I) is at the para-or meta-position of phenol hydroxyl.
In some embodiments, the compound is selected from 4-hydroxyphenylpyruvic acid (4HPPA) , 3-hydroxyphenylpropionic acid (3HPP) and 4-hydroxyphenylpropionic acid (4HPP) of which the chemical structures are shown below,
.
The drug can be administered by any suitable method known in the art, including gastrointestinal or parenteral administration. In some embodiments, the gastrointestinal administration is oral administration or nasal feeding.
The drug may be in any dosage form suitable for gastrointestinal or parenteral administration. In some embodiments, the drug is in a liquid dosage form (e.g. emulsion, microemulsion, solution, suspension, syrup or elixir) or a solid dosage form (e.g. capsule, tablet, pill, lozenge, powder or granule) for oral administration. The preferred dosage form depends on the intended mode of administration and therapeutic use.
An exemplary route of administration is oral administration. Liquid dosage form for oral administration includes pharmaceutically acceptable emulsion, microemulsion, solution, suspension, syrup, elixir and the like. In addition to the active compound, the liquid dosage form may contain an inert diluent commonly used in the art, such as water or other solvent, solubilizer and emulsifier, such as ethanol, isopropanol, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butanediol, dimethylformamide, oil (e.g., cottonseed oil, peanut oil, corn oil, germ oil, olive oil, castor oil and sesame oil) , glycerin, tetrahydrofurfuryl alcohol, polyethylene glycol, and fatty acid ester of sorbitan as well as mixtures thereof. In addition to inert diluent, liquid dosage form for oral administration may also include adjuvant, such as wetting agent, emulsifier and suspending agent, sweetening agent, flavoring agent, and fragrance. Solid dosage form for oral administration includes capsule, tablet, pill, lozenge, powder, granule and the like. In addition to the active compound, the solid dosage form may contain pharmaceutically acceptable inert excipient or carrier, such as filler (e.g., lactose, sucrose, glucose, mannitol, starch, microcrystalline cellulose, galactose, crospovidone and calcium sulfate) ; binder (e.g., carboxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and acacia) ; wetting agent (e.g., cetyl alcohol and glyceryl monostearate) ; disintegrant (e.g., agar, calcium carbonate, starch, alginic acid, sodium carboxymethyl cellulose, sodium carboxymethyl starch) ; lubricant (e.g., talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium laurel sulfate) ; and mixtures thereof.
The compound or pharmaceutical composition of the present invention can also be administered by a non-oral route.
Therefore, another exemplary route of administration is parenteral administration, for example, subcutaneous injection, intravenous injection, intraperitoneal injection, intramuscular injection, intrasternal injection, and infusion. The dosage form for parenteral administration may be an injection preparation, including injection solution, sterile powder for injection, or concentrated solution for injection. In addition to the active compound, the injection dosage form may contain a pharmaceutically acceptable carrier such as sterile water, Ringer's solution and isotonic sodium chloride solution. An appropriate additive such as antioxidant, buffer and bacteriostatic agent may also be added depending on the nature of the drug.
In addition, other carrier materials and administration methods known in the pharmaceutical field can also be used. The pharmaceutical composition of the present invention can be prepared by any well-known pharmaceutical process, such as effective formulation and administration method. The aforementioned considerations regarding effective formulation and administration method are well known in the art and described in standard textbooks. The formulation of pharmaceuticals is described in, for example, Hoover, John E., Remington's Pharmaceutical Sciences. Mack Publishing Co., Easton, Pennsylvania, 1975; Liberman et al., Pharmaceutical Dosage Forms, Marcel Decker, New York, NY, 1980; and Kibbe et al., Handbook of Pharmaceutical Excipients (3rd edition) , American Pharmaceutical Association, Washington, 1999.
In some embodiments, the drug is used for preventing and/or treating obesity, overweight, and the drug is administrated to the subject by gastrointestinal administration (e.g. oral administration) .
In some embodiments, the drug is used for preventing and/or treating nonalcoholic fatty liver disease (NAFLD) in the subject; the drug is administrated to the subject by parenteral administration; the n in formula (I) is an integer selected from 2-6.
In some embodiments, the n in formula (I) is 2 or 3.
In some embodiments, the compound is 3HPP or 4HPP.
The compound of formula (I) is found to have protective effect on the liver. Particularly, it was found that IP-injected the compound (e.g. 3HPP) protected a subject from HFD-induced hepatic steatosis. Therefore, the compound of formula (I) may serve as a drug
candidate for the prevention or therapeutics of nonalcoholic fatty liver disease (NAFLD) . In the application, NAFLD includes simple fatty liver and non-alcoholic steatohepatitis (NASH) .
The compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof may also be used in preparation of a dietary supplement, wherein the dietary supplement is used for weight management or weight control of a subject. The dietary supplement may be in a liquid dosage form (e.g. emulsion, microemulsion, solution, suspension, syrup or elixir) or a solid dosage form (e.g. capsule, tablet, pill, lozenge, powder or granule) for oral administration as described above.
In another aspect, the present application provides use of the compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in preparation of a drug or dietary supplement, wherein the drug or dietary supplement is used for modulation of intestinal immune control in a subject.
The modulation of intestinal immune control in a subject may comprises (1) inducing IgA production in the subjetct’s intestine, (2) suppressing expression of a key factor (e.g. fatty acid binding protein 2 (Fabp2) , fatty acid binding protein 4 (Fabp4) , stearoyl-CoA desaturase 1 (Scd1) , lipoprotein lipase (Lpl) and/or cluster of differentiation 36 (Cd36) ) for intestinal lipid absorption in the subject’s intestine, (3) suppressing inflammation in the subjetct’s intestine, (4) increasing MyD88 expression in the subject’s intestinal immune cells, and/or (5) increasing number of immune cells (e.g. B cells and/or Th17 cells) in the subjetct’s intestinal mucosa.
In some embodiments, the drug or dietary supplement is used for suppressing lipid absorption in the subject.
In some embodiments, the drug is used for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in the subject.
In some embodiments, the dietary supplement is used for weight management or weight control of a subject.
The compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof may be used in combination with other pharmaceutically active agents. Therefore, the drug described above may further comprises an additional pharmaceutically active agent, e.g. anti-diabetic drug, anti-obesity drug, anti-hypertensive drug, anti-atherosclerosis drug, lipid-lowering drug or anti-inflammatory drug.
In some embodiments, in the drug described above, the compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof and the additional pharmaceutically active agent are provided as separate components or mixed components. Therefore, compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof and the additional pharmaceutically active agent can be administered simultaneously, separately or sequentially.
The drug described above may comprise a "therapeutically effective amount" or "prophylactically effective amount" of compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof. The term "prophylactically effective amount" refers to an amount sufficient to prevent, stop, or delay the occurrence of a disease. The term "therapeutically effective amount" refers to an amount sufficient to cure or at least partially prevent a disease and complication thereof in a patient who has already suffered from the disease. Those skilled in the art understand that compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof may have a therapeutically effective amount varying according to the following factors: severity of disease to be treated, overall state of patient’s own immune system, patient’s general conditions such as age, weight and gender, administration mode of drug, and other treatments simultaneously administered, etc.
In some embodiments, the drug or dietary supplement comprises the compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in an amount of 0.01 mg to 10 g, e.g. 0.01 mg to 0.1 mg, 0.1 mg to 1 mg, 1 mg to 10 mg, 10 mg to 100 mg, 100 mg to 1000 mg, 1 g to 3 g, 3 g to 5 g, 5 g to 7 g, or 7 g to 10 g.
In another aspect, the present application provides a method for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in a subject, or controlling or managing weight in a subject, wherein the method comprises administrating a compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof by gastrointestinal administration (e.g. oral administration or nasal feeding) or parenteral administration to the subject; the compound isn’ t 4-hydroxyphenylacetic acid (4HPAA) in condition that the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof is parenterally administrated to the subject for preventing and/or treating NAFLD.
In some embodiments, the method is for preventing and/or treating obesity or overweight in the subject; the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal is administrated to the subject by gastrointestinal administration (e.g. oral administration) .
In some embodiments, the method is for preventing and/or treating nonalcoholic fatty liver disease (NAFLD) in the subject; the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal is administrated to the subject by parenteral administration; the n in formula (I) is an integer selected from 2-6.
In some embodiments, the method is for controlling or managing weight in the subject; the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof is administrated to the subject by gastrointestinal administration (e.g. oral administration) .
In another aspect, the present application provides a method for modulation of intestinal immune control in a subject, wherein the method comprises administrating a compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof by gastrointestinal administration (e.g. oral administration or nasal feeding) or parenteral administration to the subject.
In some embodiments, the modulation of intestinal immune control in a subject comprises (1) inducing IgA production in the subjetct’s intestine, (2) suppressing expression of a key factor (e.g. Fabp2, Fabp4, Scd1, Lpl and/or Cd36) for intestinal lipid absorption in the subject’s intestine, (3) suppressing inflammation in the subjetct’s intestine, (4) increasing MyD88 expression in the subject’s intestinal immune cells, and/or (5) increasing number of immune cells (e.g. B cells and/or Th17 cells) in the subjetct’s intestinal mucosa.
In some embodiments, the method is for suppressing lipid absorption in the subject.
In any one of the methods described above, the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof may be administrated in a liquid dosage form (e.g. emulsion, microemulsion, solution, suspension, syrup or elixir) or a solid dosage form (e.g. capsule, tablet, pill, lozenge, powder or granule) . Detailed description about the these dosage forms may be found above.
Any one of the methods described above may also comprise: administering the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof
in combination with an additional therapy. The additional therapy can be any therapy known for metabolic diseases, such as surgery, targeted therapy, immunotherapy, hormone therapy or gene therapy. This additional therapy can be administered before, simultaneously or after the administration of compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof as described herein.
In some embodiments, the additional therapy comprises administration of an additional pharmaceutically active agent selected from the group consisting of anti-diabetic drug, anti-obesity drug, anti-hypertensive drug, anti-atherosclerosis drug, lipid-lowering drug and anti-inflammatory drug.
In some embodiments, the method is used in combination with a reduced-calorie diet and/or increased physical activity.
In the present invention, the dosage regimen can be adjusted to obtain the best objective response (e.g., therapeutic or preventive response) . For example, it can be administered in a single dose, can be administered multiple times over a period of time, or the dose can be reduced or increased proportionally to the urgency of the treatment situation. In some embodiments, the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof is administrated to the subject once, twice or three times a day.
the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof has a therapeutically or prophylactically effective amount with a typical and non-limiting range of 0.01 to 500 mg/kg/day, e.g. 0.01 to 1 mg/kg/day, 1 to 20 mg/kg/day, 20 to 50 mg/kg/day, 50 to 100 mg/kg/day, 100 to 200 mg/kg/day, or 200 to 500 mg/kg/day. It should be noted that the dosage may vary with the type and severity of the symptoms to be treated. In addition, those skilled in the art understand that for any specific patient, the specific dosage regimen should be adjusted over time based on the patient’s needs and the doctor's professional evaluation; the dosage range given here is for illustrative purposes only, and does not limit the use or scope of the pharmaceutical composition of the present invention.
Definition of Terms
In the present invention, unless otherwise specified, the scientific and technical terms used herein have the meanings commonly understood by those skilled in the art. Moreover, the laboratory procedures involved herein are routine procedures widely used in the corresponding
fields. At the same time, in order to better understand the present invention, definitions and explanations of related terms are provided below.
As used herein, the term "pharmaceutically acceptable salt" refers to (i) a salt formed by an acidic functional group (e.g., -COOH) present in the compound provided by the present invention and an appropriate inorganic or organic cation (base) , which includes, but is not limited to, alkali metal salt, such as sodium salt, potassium salt, lithium salt, etc.; alkaline earth metal salt, such as calcium salt, magnesium salt, etc.; other metal salt, such as aluminum salt, iron salt, zinc salt, copper salt, nickel salt, cobalt salt, etc.; inorganic alkali salt, such as ammonium salt; organic alkali salt, such as tert-octylamine salt, dibenzylamine salt, morpholine salt, glucosamine salt, phenylglycine alkyl ester salt, ethylenediamine salt, N-methylglucamine salt, guanidine salt, diethylamine salt, triethylamine salt, dicyclohexylamine salt, N, N'-dibenzylethylenediamine salt, chloroprocaine salt, procaine salt, diethanolamine salt, N-benzyl-phenethylamine salt, piperazine salt, tetramethylammonium salt, tris (hydroxymethyl) aminomethane salt; and, (ii) a salt formed by a basic functional group (e.g., -NH2) in the compound provided by the present invention and a suitable inorganic or organic anion (acid) , which includes but is not limited to, hydrohalide salt, such as hydrofluoride salt, hydrochloride salt, hydrobromide salt, hydroiodide salt, etc.; inorganic acid salt, such as nitrate, perchlorate, sulfate, phosphate, etc.; lower alkane-sulfonate, such as methanesulfonate, trifluoromethanesulfonate, ethanesulfonate, etc.; arylsulfonate, such as benzenesulfonate, p-benzenesulfonate, etc.; organic acid salt, such as acetate, malate, fumarate, succinate, citrate, tartrate, oxalate, maleate, etc.; amino acid salt, such as glycinate, trimethylglycinate, arginine salt, ornithine salt, glutamate, aspartate, etc.
As used herein, the term "pharmaceutically acceptable ester" refers to an ester formed by -COOH present in the compound provided by the present invention and an appropriate alcohol, or an ester formed by -OH present in the compound provided by the present invention and an appropriate acid (e.g., a carboxylic acid or an oxygen-containing inorganic acid) . Suitable ester groups include, but are not limited to, formate, acetate, propionate, butyrate, acrylate, ethylsuccinate, stearyl fatty acid ester, or palmitate. In the presence of an acid or base, the ester can undergo a hydrolysis reaction to generate the corresponding acid or alcohol.
As used herein, the term "solvate" refers to a substance formed by the association of the compound of the present invention with a solvent molecule. The solvent may be an organic solvent (e.g., methanol, ethanol, propanol, acetonitrile, etc. ) , for example, the compound of the present
invention may form an ethanolate with ethanol. The compound of the present invention can also form an hydrate with water.
As used herein, the term "crystal form" refers to a crystal structure of a substance. When the substance is crystallized, due to various factors, the intra-molecular or intermolecular bonding mode changes, resulting in different arrangement of molecules or atoms in the lattice space, forming different crystal structures. The compound of the present invention may exist in one crystal structure or in multiple crystal structures, that is, have a "polymorphic form" . The compound of the present invention may exist in different crystal forms.
As used herein, the term "stereoisomer" includes conformational isomer and configurational isomer, wherein the configurational isomer mainly includes cis-trans isomers and optical isomers. The compound of the present invention may exist in the form of stereoisomer, and therefore encompass all possible stereoisomeric forms, and any combination or any mixture thereof, for example, a single enantiomer, a single diastereomer or a mixture of the above. When the compound of the present invention comprises an alkene double bond, unless otherwise specified, it includes cis isomer and trans isomer, and any combination thereof.
As used herein, the term "pharmaceutically acceptable carrier or excipient" refers to a carrier and/or excipient that is pharmacologically and/or physiologically compatible with the subject and the active ingredient, which is well known in the art (see, for example, Remington's Pharmaceutical Sciences. Edited by Gennaro AR, 19th ed. Pennsylvania: Mack Publishing Company, 1995) , and includes, but is not limited to: disintegrant, binder, surfactant, glidant, lubricant, pH adjuster, ionic strength enhancer, agent for maintaining osmotic pressure, agent for delaying absorption, diluent, antioxidant, coloring agent, flavoring agent, preservative, taste masking agent, etc. For example, non-limiting examples of disintegrant include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methylcellulose, microcrystalline cellulose, lower alkyl-substituted hydroxypropylcellulose, starch, pregelatinized starch and sodium alginate. Non-limiting examples of binder include microcrystalline cellulose, gelatin, sugar, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinized starch, hydroxypropyl cellulose, and hydroxypropyl methyl cellulose. Non-limiting examples of diluent include lactose (monohydrate, spray-dried monohydrate, anhydrous, etc. ) , mannitol, xylitol, glucose, sucrose, sorbitol, microcrystalline cellulose, starch, and calcium hydrogen phosphorate dihydrate. Non-
limiting examples of surfactant include sodium lauryl sulfate and polysorbate 80. Non-limiting examples of glidant include silica and talc. Non-limiting examples of lubricant include magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate and sodium lauryl sulfate. Non-limiting example of pH adjusting agent includes but is not limited to phosphate buffer. Ionic strength enhancer includes, but is not limited to, sodium chloride. Agent for maintaining osmotic pressure includes but is not limited to sugar, NaCl and the like. Agent that delays absorption includes, but is not limited to, monostearate and gelatin. Preservative includes, but is not limited to, various antibacterial agents and antifungal agents, such as thimerosal, 2-phenoxyethanol, paraben, chlorobutanol, phenol, sorbic acid and the like.
As used herein, the term "prevention" refers to a method performed to prevent or delay the occurrence of a disease or disorder or symptom in a subject. As used herein, the term "treatment" refers to a method performed to obtain a beneficial or desired clinical result. For the purpose of the present invention, the beneficial or desired clinical result includes, but is not limited to, alleviating symptom, narrowing disease scope, stabilizing (i.e., not getting worse) disease state, delaying or slowing disease development, improving or alleviating disease state, and relieving symptom (whether partially or fully) , whether detectable or undetectable. In addition, "treatment" may also refer to that a survival period is prolonged compared to an expected survival period (if not receiving treatment) .
As used herein, the term "subject" refers to a mammal, such as a primate mammal, such as a human.
Figure 1 Oral intake of 4HPAA, 3HPP, and 4HPP suppress weight gain in HFD-fed mice. (A) Schematic design of oral treatment in the HFD mouse model. (B) A presentative image of ND-fed, HFD-fed, and HFD-fed plus 4HPAA-treated mice. (C) The weight gain of ND-fed and HFD-fed mice treated with or without 4HPAA 8 weeks was measured and plotted on the chart. (D) The body fat percentage of ND-fed and HFD-fed mice treated with or without 4HPAA for 8 weeks was measured and plotted on the chart. (E) The food intake of HFD-fed mice treated with or without 4HPAA after 8 weeks was measured and plotted on the chart. (F) The energy expenditure of HFD-fed mice treated with or without 4HPAA after 8 weeks. (n=5 mice per group, ****P< 0.0001, unpaired Student’s t-test. ) (G) Structural formulas of 4-HPAA, 3HPP, 4-HPP, and
tyrosol. (H) Body weight growth curve of ND/HFD-fed mice treated with or without 4HPAA, 4HPP, 3HPP, and tyrosol in drinking water. (I) Body weight gain of ND/HFD-fed mice orally treated with or without 4HPAA, 4HPP, 3HPP, and tyrosol for 10 weeks. (J) Body fat percentage of ND/HFD-fed mice orally treated with or without 4HPAA, 4HPP, 3HPP, and tyrosol for 10 weeks. For (C-E and H-J) , error bars indicate mean±s.e.m., statistical analysis was performed using the two-tailed Mann-Whitney test, ns=not significant, *p<0.05, **p<0.01, ***p<0.001, ****p< 0.0001.
Figure 2 Metabolic performances of the HFD-fed mice orally treated with or without 4HPAA. (A) Daily food intake of these mice during the treatment. (B) Whole-body oxygen consumption (upper panel) and carbon dioxide production (lower panel) after 8 weeks (n=5 mice per group) .
Figure 3 4HPAA intervention blocks the body weight gain in mice already become obese. (A) Schematic view of the experiment design. Two groups of mice were fed with HFD under the same condition for 14 weeks. After 8 weeks, one group was kept given normal water (control) and the other was changed to 4HPAA-containing water for the next 6 weeks. The mouse's body weight was measured weekly. (B) The average body weights of these two groups during 8-14 weeks are shown. n=15 mice per group, error bars indicate mean ± s.e.m., *p < 0.05, two-tailed Mann-Whitney test.
Figure 4 Similar fecal metabolome was observed in 4HPAA, 4HPP, and 3HPP treatments. (A) The Venn diagram shows that the upregulated metabolites in the 4HPAA, 4HPP, and 3HPP groups are largely overlapped. (B) The heat map shows the overlapped upregulated metabolites in (A) .
Figure 5 4HPAA, 3HPP, and 4HPP alleviate adipocyte hypertrophy and hepatic steatosis. (A) Representative images of H&E stained adipose tissues, including iBAT, iWAT, and eWAT, from ND/HFD-fed mice orally treated with or without 4HPAA, 4HPP, 3HPP, and tyrosol after 10 weeks. (B-D) The average diameters of adipocytes in iBAT (B) , iWAT (C) , and eWAT (D) were measured and plotted on the charts. (E) Representative images of H&E stained liver sections from ND/HFD-fed mice orally treated with or without 4HPAA, 4HPP, 3HPP, and tyrosol after 10 weeks. (F) The fatty liver histological scores in (E) were assessed and plotted on the diagram. For (B-D and F) , error bars indicate mean±s.e.m., statistical analysis was performed using two-tailed Mann-Whitney test; ns=not significant, *p<0.05, **p<0.01, ***p<0.001, ****p< 0.0001.
Figure 6 Serum biomarkers measured in ND/HFD-fed mice orally treated with or without 4HPAA, 3HPP, and 4HPP. Error bars indicate mean±s.e.m., ns: not significant, *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, two-tailed Mann-Whitney test.
Figure 7 IP injected 4HPAA and 3HPP failed to inhibit body fat accumulation in HFD-fed mice. (A) Schematic design of IP treatment of 4HPAA/3HPP in the HFD mouse model. (B-C) ND/HFD-fed mice were IP injected with or without 4HPAA and 3HPP twice a week. After 12 weeks, the body weight gain (B) and fat percentage (C) of these mice were assessed and plotted on the charts. (D) Representative images of H&E stained adipose tissues, including iBAT, iWAT, and eWAT, from ND/HFD-fed mice IP injected with saline, 4HPAA, and 3HPP for 12 weeks. (E) Representative images of H&E stained liver sections from ND/HFD-fed mice IP injected with saline, 4HPAA, and 3HPP after 12 weeks. (F) The fatty liver histological scores in (E) were assessed and plotted on the diagram. For (B-C and F) , error bars indicate mean±s.e.m., ns=not significant, *p<0.05, two-tailed Mann-Whitney test.
Figure 8 4HPAA blocks lipid absorption and induces IgA production in the gut. (A) Volcano plot of colon transcripts. 63 upregulated gene hits and 173 downregulated gene hits (with foldchange over 2 and p-value less than 0.05) were highlighted in red and blue respectively. (B) Gene set enrichment analysis based on the upregulated and downregulated gene hits. (C) The heat map displays 25 top-upregulated genes, which all encode immunoglobulins, in the 4HPAA-treated HFD-fed mice compared with control HFD-fed mice. (D) The heat map displays all downregulated genes. Notable genes related to lipid absorption and metabolism are marked with blue labels. (E) RT-PCR analysis shows that the transcription of Cd36 and Scd1 was drastically inhibited in the 4HPAA-fed mice compared to control mice. (F) IHC analysis shows the reduced protein level of Cd36 in the 4HPAA-fed mice compared to control mice. (G) Immunofluorescence analysis shows increased mucosa infiltration of B cells (anti-IgA, upper panel) and Th17 cells (anti-IL17, lower panel) in HFD-fed mice orally treated with 4HPAA or 3HPP. (G) IHC analysis shows increased Myd88 expression in immune cells (marked by red arrows) of HFD-fed mice orally treated with 4HPAA.
Figure 9 Behavior tests for ND-fed mice treated with or without 4HPAA. No overt behavioral differences were observed in the elevated plus maze (A) , open field test (B) , and Y-maze (C) . n=5 mice per group, error bars indicate mean ± s.e.m., ns: not significant, statistical analysis was assessed by two-tailed Mann-Whitney test.
Figure 10 Schematic drawing of the proposed model for immune control by microbial AAA metabolites in the intestinal epithelium.
Specific Models for Carrying Out the Present Invention
The embodiments of the present invention will be described in detail below in conjunction with examples, but those skilled in the art will understand that the following examples are only used to illustrate the present invention and should not be regarded as limiting the scope of the present invention.
Unless otherwise specified, the experiments and methods described in the examples are basically performed according to conventional methods well known in the art and described in various references. If specific conditions are not indicated in the examples, it shall be carried out in accordance with conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used without the manufacturer's indication are all conventional products that are commercially available. Those skilled in the art know that the examples describe the present invention by way of example and are not intended to limit the scope of protection claimed by the present invention. All publications and other references mentioned in the context are incorporated in their entirety by reference.
Materials
Mouse feed, reagents, and antibodies were purchased from the commercial vendors: High Fat Diet (OpenSource Diets, #D12492) , Normal Diet (OpenSource Diets, #D12450J) , Brain Heart Infusion (Solarbio, Cat#B8130) , 4HPAA (Sigma-Aldrich, Cat#H50004) , 3HPP (Leyan, Cat#1062804) , 4HPP (Leyan, Cat#1033824) , tyrosol (Cayman, Cat#27600) , acetonitrile (J&K, Cat#75-05-8) , Antifade Mounting Medium with DAPI (Beyotime, Cat#P0131) , rabbit monoclonal antibodies against MyD88 (GeneTex, Cat#GTX112987) and Cd36 (ABclonal, Cat#A19016) , FITC labeled rabbit monoclonal antibody against IgA (Biolegend, Cat#1165-02) , Fluor 488 labeled rabbit monoclonal antibody against IL-17 (Biolegend, Cat#506910) .
Mice
Specific pathogen free (SPF) grade C57BL/6J mice (male, aged 6-8 weeks) were purchased from Laboratory Animal Resources Center at Westlake University (Hangzhou, China) . All mice housed at 20–24 ℃ with 40–60%humidity and had a 12 h cycle of
light/darkness (7 a.m. to 7 p.m. ) . All animal protocols were approved by the Institutional Animal Care and Use Committee (IACUC) of Westlake University.
Methods
(1) Animal Experiments
Mice were fed a HFD (60%fat) or ND (10%fat) . For oral delivery, drinking water with 1.5 mg/ml 4HPAA, 3HPP, 4HPP, or tyrosol was supplemented (200 mg/kg/d) for 8-10 weeks. For IP injection, the dosage of 4HPAA or 3HPP was a series of gradient diluted concentrations: 8, 2.8, 0.9 and 0.3 mg/kg, each of which were intraperitoneal injected twice a week for 8 weeks. In this study, mouse body weight was measured at the beginning and weekly afterwards. At specific time points, fresh fecal samples were collected and stored at -80℃. Fat mass values were assessed at the end of each experiment using EchoMRI 100H Body Composition Analyzer (EchoMRI, USA) . Serum sample of each mouse was collected at the terminal experiment and freshly detected (HITACHI Automatic Aralyzer, Cat#3100) for the biochemical items mentioned in this study, including AST (aspartate transaminase) , ALT (alanine transaminase) , LDL-C (low-density lipoprotein cholesterol) , HDL-C (high-density lipoprotein cholesterol) , and TC (total cholesterol) .
(2) IHC, H&E, and IF staining of mouse tissues and histopathological analysis
Mice were sacrificed and their liver, colon, and adipose tissues were fixed in formalin for 12 hours, dehydrated with gradient alcohol, cleared with xylene, and embedded in paraffin. Paraffin blocks were cut into 5 μm thick sections and either stained with H&E or detected with specific antibodies. Cd36 and MyD88 (anti-Cd36: 1: 200; anti-MyD88: 1: 500) were detected by immunochemistry analysis and IgA-positive B cells and IL-17-positive T cells (anti-IgA: 1: 400; anti-IL-17: 1: 400) were detected by immunofluorescence analysis. The images were captured using either an upright Nikon Motorized Fluorescence Microscope (for H&E and IHC) or Nikon Spinning-disk Confocal Microscope (for IF) . The pathology was assessed and scored by two pathologists blinded on a scale of 0 to 4 (normal, minimal, mild, moderate, and severe) .
(3) Fecal microbiome analysis
To characterize the mice fecal microbiota communities, fecal DNA samples were extracted using thesoil DNA kit (Omega Bio-Tek, Norcross, GA, U.S. ) , according to manufacturer instructions. Using the extracted DNA as a template, the
upstream primer 338F (5 '-actCCTACGGgaggCAGCAGCAG-3') carrying barcode sequence and downstream primer 806R (5 '-GGACTACHVGGGTWTCTAAT-3') were used to target the V3-V4 variable region of 16S rRNA gene. After 16S rDNA amplification, the NEXTFLEX Rapid DNA-Seq Kit was used to construct the library of the purified PCR products, and sequencing was performed using Illumina's Miseq PE300/NovaSeq PE250 platform (Shanghai Majorbio Bio-pharm Biotechnology Co., Ltd. ) . Fastq-files were demultiplexed by the MiSeq Controller Software (Bcl2fastq version 2.20, Illumina Inc. ) . The sequences were trimmed for amplification primers, and sequencing adapters, merge-paired, and quality filtered by Quantitative Insights into Microbial Ecology (QIIME) software version 2-2020.2. DADA2 was used for amplicon sequence variants (ASVs) clustering equaling 100%. A representative sequence was picked for each ASV and the Sliva reference database version 138 was used to annotate taxonomic information. The absolute abundance table was extracted from the pipeline and converted to relative abundances by normalizing for analyzing the composition of gut microbiota by QIIME2.
(4) RNA-seq analysis
Mice were sacrificed after 3 months of the oral treatment and freshly collected colon tissue sections (50-100 mg) were quick-frozen and stored in liquid nitrogen. Following RNA purification, reverse transcription, library construction, and sequencing were performed at Shanghai Majorbio Bio-pharm Biotechnology Co., Ltd. (Shanghai, China) . Differential expression analysis: based on the expression of quantitative results, analysis of genetic variations between groups, gain occurred between the two groups of differentially expressed genes, variance analysis software for DESeq2 (3.16) , the threshold value is: | log2FC | > = 1 &p-value < 0.05.
(5) Reverse transcription and real-time qPCR
Colon tissues from sacrificed mice after 3 months of the oral drug trial were dissected to 50-100mg and were slightly washed with 0.9%saline. Total RNA isolation of mice colon tissues has followed the manuscript of a MolPure Cell/Tissue Total RNA Kit (Yeasen, China) . Reverse transcription was performed using an ABScript III RT Master Mix for qPCR with gDNA Remover (ABclonal, Cat#RK20429) from 2 μg of total RNA. The real-time PCR of Cd36 (cd36_RT-F: TCATATTGTGCTTGCAAATCCA, cd36_RT-R: TGTAGATCGGCTTTACCAAAGATG) , Scd1 (scd1_RT-F:
CTTCTTCTCTCACGTGGGTTG, scd1_RT-R: CGGGCTTGTAGTACCTCCTC) , and Gapdh (Gapdh_RT-F: CTCACGGCAAATTCAACG, Gapdh_RT-R: GACTCCACGACATACTCAG) were performed and the relative count of genes was calculated by normalizing to Gapdh mRNA. Universal SYBR Green Fast qPCR Mix (ABclonal, Cat #RK21203) and Jena Qtower3G instrument (Jena, Germany) conducted qPCR experiments.
(6) Statistical analysis
The associations between 4HPAA and blood lipid parameters (TG, TC, LDL, and HDL) were examined using a multivariable linear regression, adjusted for the same covariates as the above model. Unpaired Student’s t-test or Mann-Whitney test was used for comparisons between two groups. For multi-group comparisons, an analysis of variance tests followed by Tukey’s tests between groups was performed. All tests were two-tailed and a p-value of <0.05 was considered statistically significant. Data were plotted and analyzed using R version 3.6.3 and GraphPad Prism (version 9.0) .
Example 1 Evaluation of the effect of oral intake of 4HPAA on obesity
Mice were fed with either a normal diet (ND) or HFD and routinely given water (control) or water containing 1.5 mg/ml (~10 mM) 4HPAA for 8 weeks (Figure 1A) . The average body weight of 4HPAA-treated HFD-fed mice was drastically lower than the non-treated HFD-fed mice and similar to the ND-fed mice (Figure 1B) . When comparing the body weight gain, 4HPAA-treated HFD-fed mice (~7.90 g) were close to the ND-fed mice either treated (~6.40 g) or non-treated (~6.64 g) with 4HPAA, but is ~45%less than the non-treated HFD-fed mice (~14.31 g) (Figure 1C) . Consistent with the body weight gain, the fat percentage of 4HPAA-treated HFD-fed mice (~23.6%) is also much lower than non-treated HFD-fed mice (~36.1%) (Figure 1D) . Interestingly, if mice had already been fed with HFD for 8 weeks and become obese, giving 4HPAA afterward could still reduce the subsequent weight gain (Figure 2) , indicating that 4HPAA protects mice from fat accumulation, regardless of the progression stage of obesity.
During the HFD feeding, the food intake of control mice gradually reduced as they became obese, while 4HPAA-treated mice kept a relatively steady food consumption (Figure 3A) . After three months, 4HPAA-fed mice ate twice the amount of daily food compared to the control group (Figure 1E) . On the other hand, 4HPAA-fed mice had only ~10%higher metabolic expenditure than the obese control mice (Figure 1F and Figure 3B) .
Besides, 4HPAA-feeding did not affect the body weight gain and fat percentage in ND-fed mice (Figures 1C-D) . A series of behavior tests were further performed, including the elevated plus maze, open field test, and Y-maze to monitor the potential pharmacological effects on the nervous system. No obvious difference was observed between ND-fed mice treated with or without 4HPAA (Figure 3) , suggesting that oral delivery of 4HPAA at millimolar concentration has no overt pharmacotoxicity to the mice.
Example 2 Evaluation of the effect of oral intake of 3HPP or 4HPP on obesity
The ability of other metabolites in the Phe-Tyr metabolic pathways in suppressing HFD-induced weight gain was tested. Three structurally related phenolic hydroxyl compounds, 4-hydroxyphenylpropionic acid (4HPP) , 3-hydroxyphenylpropionic acid (3HPP) , and tyrosol, were selected (Figure 1G) . 4HPAA, 4HPP, 3HPP, or tyrosol were added to the drinking water at a concentration of 1.5 mg/mL. Oral intake of 4HPP and 3HPP, but not tyrosol, effectively reduced body weight gain and fat accumulation in HFD-fed mice (Figures 1H-J) . It seems that molecules with shared structural features containing a phenolic hydroxyl group on one side and a carboxyl group on the other terminal would have the bioactivity to suppress body weight gain in HFD-fed mice.
Feces from the above mice in the final week were collected. The fecal metabolomics analysis showed similar intestinal metabolic environments of the HFD-fed mice orally treated with 4HPAA, 3HPP, and 4HPP, while the metabolome patterns of the control and tyrosol-treated mice were close (Figure 4) . These data imply that 4HPAA, 3HPP, or 4HPP may exert the protection role against obesity in the same manner.
Example 3 Evaluation of the effect of 4HPAA, 3HPP and 4HPP on adipocyte hypertrophy and hepatic steatosis
As fat accumulation was drastically inhibited by 4HPAA, 3HPP, and 4HPP treatment, the effects of these molecules on different adipose tissues were assessed by histological analysis. Interscapular brown adipose tissue (iBAT) , inguinal white adipose tissue (iWAT) , and epididymal white adipose tissue (eWAT) were dissected from those HFD-fed mice, sectioned into slides, stained with hematoxylin and eosin (H&E) , and examed under the microscopy. As expected, adipocytes in the iBAT, iWAT, and eWAT from the HFD-fed mice were larger compared to the ones from the ND-fed mice (Figures 5A-D) . Such adipocyte hypertrophy is a typical phenotype of obesity. In contrast, adipocytes from the HFD-fed mice treated with
4HPAA, 3HPP, and 4HPP showed smaller sizes, which is more similar to the ND-fed mice. Oral feeding of tyrosol failed to rescue adipocyte hypertrophy in the HFD-fed mice (Figures 5A-D) . Interestingly, adipocytes in the iBAT from 4HPAA, 3HPP, or 4HPP-treated HFD-fed mice were even slightly smaller than the ones from the ND-fed mice. The brown adipocytes might be more sensitive upon 4HPAA, 3HPP, or 4HPP treatment.
HFD-induced obesity is normally accompanied by the nonalcoholic fatty liver, also called hepatic steatosis. By histological analysis of the liver sections of these mice, it was found that 4HPAA, 3HPP, and 4HPP feeding all effectively alleviated the hepatic steatosis induced by HFD, while tyrosol had no protective effects (Figures 5E-F) . Classic serum biomarkers for liver injury and blood lipid disorder were further measured, including ALT, AST, LDL, HDL, and TC. The 4HPAA, 3HPP, or 4HPP-treated HFD-fed mice had lower TC and LDL compared to the control HFD-fed. No obvious difference was observed for ALT, AST, and HDL (Figure 6) .
Saline with or without 12 mg/kg 4HPAA and 3HPP was intraperitoneally (IP) injected into the HFD-fed mice twice a week (Figure 7A) . It was found that IP-injected 4HPAA slightly protected the liver, while IP-injected 3HPP could better alleviate the hepatic steatosis (Figures 7E-F) . These data suggest that high concentrations of injected 4HPAA/3HPP (~20 mM) may target the liver and reduce hepatic steatosis.
Example 4 Evaluation of the effect of 4HPAA on lipid absorption and IgA production in the intestine
To understand how these AAA metabolites protest against obesity through the intestinal epithelium, the transcriptomes of intestinal tissue from the HFD-fed mice with or without 4HPAA feeding were compared. RNA-sequencing (RNA-seq) revealed 63 upregulated genes and 173 downregulated genes in the colon tissue with a fold-change over 2 (Figure 8A and Figure 9) . Gene set enrichment analysis (GSEA) showed that the B cell-mediated immune network for IgA production was mostly enriched, whereas pathways involved in lipid metabolism, including monocarboxylic acid catabolism, fatty acid catabolism, triglyceride metabolism, fatty acid metabolism, and lipid oxidation were strongly suppressed (Figure 8B) . Nearly all top-upregulated genes directly encode immunoglobulins in HPAA-fed mice (Figure 8C and Figure 9) . The immunofluorescence analysis further confirmed that the HPAA-fed mice had a higher IgA level in the colonic epithelium compared to the control mice (Figure 8D) . On the other hand, multiple genes that function in lipid absorption and metabolism, including but not limited to
Fabp2, Fabp4, Scd1, Lpl, and Cd36, were largely suppressed in HPAA-fed mice (Figure 8C) .
The decreased transcription of Cd36 and Scd1 in the HPAA-fed mice was validated by reverse transcription PCR (Figure 8E) . Notably, Cd36 is a key protein that transports dietary fatty acids into cells and modulates proinflammatory response. The reduced protein level of Cd36 in HPAA-fed mice was further validated by immunohistochemistry analysis (Figure 8F) . Because RNA-seq showed that IgA production was drastically increased upon 4HPAA treatment, B cells in the mouse intestine were next stained using an anti-IgA antibody. Immunofluorescence analysis showed increased B cells in the mucosa layer of mice treated with 4HPAA or 3HPP (Figure 8G) , which is consistent with the RNA-seq result. It was recently reported that Cd36 can be directly inhibited by IL-17. By using an anti-IL17 antibody, it was also found that mucosa infiltration of Th17 cells in mice treated with 4HPAA or 3HPP increased (Figure 8G) , suggesting intestinal T cells are also regulated by these metabolites. Interestingly, previous studies showed that the aging T-Myd88-/-mice would become obese and those mice had very little gut IgA and increased Cd36 expression. To test whether the MyD88 pathway is involved, MyD88 levels in the control and HPAA-fed mice were measured and it was found that MyD88 expression in the resident intestinal immune cells increased (Figure 8H) . Taken together, it was propose that AAA metabolites such as 4HPAA and 3HPP regulate B and T cell-mediated intestinal immune control to affect host lipid absorption and metabolism (Figure 10) .
Although the specific embodiments of the present invention have been described in detail, those skilled in the art will understand that various modifications and substitutions can be made to those details according to all the teachings that have been disclosed, and these changes are all within the protection scope of the present invention. The full scope of the present invention is given by the appended claims and any equivalents thereof.
Claims (20)
- Use of a compound of formula (I) , pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof in preparation of a drug, wherein the drug is used for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in a subject; the drug is administrated to the subject by gastrointestinal or parenteral administration;
wherein n is an integer selected from 1-6;the compound isn’t 4-hydroxyphenylacetic acid (4HPAA) in condition that the compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof is parenterally administrated to the subject for preventing and/or treating NAFLD. - The use of claim 1, wherein the n in formula (I) is 1 or 2.
- The use of claim 1 or 2, wherein - (CH2) n-COOH in formula (I) is at the para-or meta-position of phenol hydroxyl.
- The use of any one of claims 1-3, wherein the compound is selected from 4-hydroxyphenylpyruvic acid (4HPPA) , 3-hydroxyphenylpropionic acid (3HPP) and 4-hydroxyphenylpropionic acid (4HPP) of which the chemical structures are shown below,
- The use of any one of claims 1-4, wherein the gastrointestinal administration is oral administration or nasal feeding.
- The use of any one of claims 1-5, wherein the drug is in a liquid dosage form (e.g. emulsion, microemulsion, solution, suspension, syrup or elixir) or a solid dosage form (e.g. capsule, tablet, pill, lozenge, powder or granule) for oral administration.
- The use of any one of claims 1-6, wherein the drug is used for preventing and/or treating obesity or overweight in the subject; the drug is administrated to the subject by gastrointestinal administration (e.g. oral administration) .
- The use of any one of claims 1-6, wherein the drug is used for preventing and/or treating nonalcoholic fatty liver disease (NAFLD) in the subject; the drug is administrated to the subject by parenteral administration; the n in formula (I) is an integer selected from 2-6.
- The use of claim 8, wherein the n in formula (I) is 2 or 3.
- The use of claim 8 or 9, wherein the compound is 3HPP or 4HPP.
- The use of any one of claims 1-10, wherein the nonalcoholic fatty liver disease (NAFLD) is simple fatty liver or non-alcoholic steatohepatitis (NASH) .
- Use of a compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof defined in any one of claims 1-4 in preparation of a dietary supplement, wherein the dietary supplement is used for weight management or weight control of a subject.
- The use of claim 12, wherein the dietary supplement is in a liquid dosage form (e.g. emulsion, microemulsion, solution, suspension, syrup or elixir) or a solid dosage form (e.g. capsule, tablet, pill, lozenge, powder or granule) for oral administration.
- Use of a compound, pharmaceutically acceptable salt or ester, stereoisomer, hydrate, solvate or crystal form thereof defined in any one of claims 1-4 in preparation of a drug or dietary supplement, wherein the drug or dietary supplement is used for modulation of intestinal immune control in a subject.
- The use of claim 14, wherein the modulation of intestinal immune control in a subject comprises (1) inducing IgA production in the subjetct’s intestine, (2) suppressing expression of a key factor (e.g. Fabp2, Fabp4, Scd1, Lpl and/or Cd36) for intestinal lipid absorption in the subject’s intestine, (3) suppressing inflammation in the subjetct’s intestine, (4) increasing MyD88 expression in the subject’s intestinal immune cells, and/or (5) increasing number of immune cells (e.g. B cells and/or Th17 cells) in the subjetct’s intestinal mucosa.
- The use of claim 14 or 15, wherein the drug or dietary supplement is used for suppressing lipid absorption in the subject.
- The use of any one of claims 14-16, wherein the drug is used for preventing and/or treating obesity, overweight or nonalcoholic fatty liver disease (NAFLD) in the subject.
- The use of any one of claims 14-16, wherein the dietary supplement is used for weight management or weight control of a subject.
- The use of any one of claims 1-11 and 14-17, wherein the drug further comprises an additional pharmaceutically active agent.
- The use of claim 19, wherein the additional pharmaceutically active agent is selected from the group consisting of anti-diabetic drug, anti-obesity drug, anti-hypertensive drug, anti-atherosclerosis drug, lipid-lowering drug and anti-inflammatory drug.
Priority Applications (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| PCT/CN2023/078793 WO2024178621A1 (en) | 2023-02-28 | 2023-02-28 | Use of compounds in preventing and/or treating obesity or nafld |
| CN202380082492.0A CN120435287A (en) | 2023-02-28 | 2023-02-28 | Use of compounds in preventing and/or treating obesity or NAFLD |
Applications Claiming Priority (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| PCT/CN2023/078793 WO2024178621A1 (en) | 2023-02-28 | 2023-02-28 | Use of compounds in preventing and/or treating obesity or nafld |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024178621A1 true WO2024178621A1 (en) | 2024-09-06 |
Family
ID=92589269
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2023/078793 Ceased WO2024178621A1 (en) | 2023-02-28 | 2023-02-28 | Use of compounds in preventing and/or treating obesity or nafld |
Country Status (2)
| Country | Link |
|---|---|
| CN (1) | CN120435287A (en) |
| WO (1) | WO2024178621A1 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN119185276A (en) * | 2024-09-30 | 2024-12-27 | 徐州医科大学 | Application of deaminated tyrosine in preparing medicament for treating cardiomyopathy |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20100104548A1 (en) * | 2005-06-24 | 2010-04-29 | Albert Einstein College Of Medicine Of Yeshiva University | Modulation of amino acid metabolism in the hypothalamus |
| CN109364056A (en) * | 2018-09-25 | 2019-02-22 | 浙江欧谱生物科技有限公司 | Application of the g protein coupled receptor 109A inhibitor in medicine preparation |
| EP3466417A1 (en) * | 2017-10-04 | 2019-04-10 | Sorbonne Université | Compounds for the prevention and treatment of glucose intolerance related conditions and obesity |
| CN110123827A (en) * | 2019-06-17 | 2019-08-16 | 中南民族大学 | A kind of pharmaceutical composition and its preparation method and application treated by metabolic disorder associated diseases |
-
2023
- 2023-02-28 WO PCT/CN2023/078793 patent/WO2024178621A1/en not_active Ceased
- 2023-02-28 CN CN202380082492.0A patent/CN120435287A/en active Pending
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20100104548A1 (en) * | 2005-06-24 | 2010-04-29 | Albert Einstein College Of Medicine Of Yeshiva University | Modulation of amino acid metabolism in the hypothalamus |
| EP3466417A1 (en) * | 2017-10-04 | 2019-04-10 | Sorbonne Université | Compounds for the prevention and treatment of glucose intolerance related conditions and obesity |
| CN109364056A (en) * | 2018-09-25 | 2019-02-22 | 浙江欧谱生物科技有限公司 | Application of the g protein coupled receptor 109A inhibitor in medicine preparation |
| CN110123827A (en) * | 2019-06-17 | 2019-08-16 | 中南民族大学 | A kind of pharmaceutical composition and its preparation method and application treated by metabolic disorder associated diseases |
Non-Patent Citations (1)
| Title |
|---|
| GUO JINGLING, WANG PAN, CUI YIFAN, HU XIAOSONG, CHEN FANG, MA CHEN: "Protective Effects of Hydroxyphenyl Propionic Acids on Lipid Metabolism and Gut Microbiota in Mice Fed a High-Fat Diet", NUTRIENTS, vol. 15, no. 4, 20 February 2023 (2023-02-20), XP093206022, DOI: 10.3390/nu15041043 * |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN119185276A (en) * | 2024-09-30 | 2024-12-27 | 徐州医科大学 | Application of deaminated tyrosine in preparing medicament for treating cardiomyopathy |
Also Published As
| Publication number | Publication date |
|---|---|
| CN120435287A (en) | 2025-08-05 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US12403114B2 (en) | Cannabinoid acid ester compositions and uses thereof | |
| EP3458448B1 (en) | Fasn inhibitors for use in treating non-alcoholic steatohepatitis | |
| CN101843904A (en) | Methods of preventing or delaying the manifestation of cancer using artemisinin-like compounds | |
| JP2023525571A (en) | Concomitant treatment of liver damage | |
| JPWO2019160057A1 (en) | Prophylactic or therapeutic agents and pharmaceutical compositions for inflammatory or bone diseases | |
| JP2023525570A (en) | Concomitant treatment of liver damage | |
| WO2024178621A1 (en) | Use of compounds in preventing and/or treating obesity or nafld | |
| TW201609094A (en) | Novel methods for treating cancer | |
| JP2024531374A (en) | Taxol conjugate compounds, pharmaceutical compositions containing same and methods of using same - Patents.com | |
| US9717718B2 (en) | Anti-inflammatory compounds | |
| JP2005539091A (en) | Growth hormone releasing peptides in the treatment or prevention of atherosclerosis and hypercholesterolemia | |
| Qi et al. | Ginseng-derived GABAFG ameliorates type 2 diabetes mellitus by modulating autophagy-lysosome pathway and gut microbiota | |
| US9314438B2 (en) | Elevation of the plasma HDL-cholesterol level | |
| US8093210B2 (en) | Treatment of sepsis with 5-ethyl-1-phenyl-2(1H)-pyridone | |
| Ma et al. | Damascenone inhibits osteoclastogenesis by epigenetically modulating Nrf2-mediated ROS scavenge and counteracts OVX-induced osteoporosis | |
| CN107224440B (en) | A kind of CCR2 antagonist and use thereof | |
| US20120029031A1 (en) | Metadoxine for use as inhibitor of hepatic fibrosis | |
| CN116115760A (en) | Application of EED inhibitor in preparation of medicine for treating neuroimmune diseases | |
| US20230027192A1 (en) | Methods of using soluble solid dispersions of rifaximin | |
| CA3013666C (en) | Use of stiripentol and their derivatives for decreasing urinary oxalate concentration in an individual | |
| WO2005055997A1 (en) | Medicinal composition for treating and preventing inflammatory disease | |
| Chang et al. | A Novel Polymorphic Form of Sodium Benzoate (Ω-NaBen): Improved Solubility, Stability, Central Nervous System Effects, and Antipsychotic Activities via D-Amino Acid Regulation | |
| HK40063789B (en) | Cannabinoid acid ester compositions and uses thereof | |
| Jiang et al. | Clinical efficacy of oxycontin combined with gabapentin on neuropathic cancer pain and its effect on immune function | |
| Wang et al. | Oct-B: A derivative of L-BAIBA significantly alleviating high-fat diet-induced obesity in mice |
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: 23924593 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 202380082492.0 Country of ref document: CN |
|
| WWP | Wipo information: published in national office |
Ref document number: 202380082492.0 Country of ref document: CN |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |