WO2009143763A1 - 丹参酮iia在制备治疗肥胖及胰岛素抵抗的药物中的应用 - Google Patents

丹参酮iia在制备治疗肥胖及胰岛素抵抗的药物中的应用 Download PDF

Info

Publication number
WO2009143763A1
WO2009143763A1 PCT/CN2009/071970 CN2009071970W WO2009143763A1 WO 2009143763 A1 WO2009143763 A1 WO 2009143763A1 CN 2009071970 W CN2009071970 W CN 2009071970W WO 2009143763 A1 WO2009143763 A1 WO 2009143763A1
Authority
WO
WIPO (PCT)
Prior art keywords
tanshinone
oxime
tanshinone oxime
composition
ppar
Prior art date
Application number
PCT/CN2009/071970
Other languages
English (en)
French (fr)
Inventor
秦莹
宫振伟
Original Assignee
中国科学院上海生命科学研究院
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 中国科学院上海生命科学研究院 filed Critical 中国科学院上海生命科学研究院
Publication of WO2009143763A1 publication Critical patent/WO2009143763A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/53Lamiaceae or Labiatae (Mint family), e.g. thyme, rosemary or lavender
    • A61K36/537Salvia (sage)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones

Definitions

  • the present invention relates to the field of biotechnology and medicine, and in particular to the use of tanshinone oxime in the prevention, control or treatment of obesity, insulin resistance and diabetes. Background technique
  • PPAR ⁇ peroxisome proliferator-activated receptor Y
  • RXR xanthene X receptor
  • PPRE PPAR reflecting element
  • PPAR gamma plays an important role in adipocyte development, thus making PPAR gamma a metabolic disease-related drug target over the past few decades.
  • thiazolidinediones anti-diabetic drugs (TZDs;), pioglitazone, rosiglitazone are known agonists of ⁇ , and their insulin sensitizing effects are mediated through their pleiotropic effects in adipose tissue.
  • Agonists of PPARj such as TZDs, are currently the first line of treatment for type 2 diabetes.
  • these drugs have many side effects, including weight gain, stagnant water, pulmonary edema, and macular edema, which in turn increase the risk of congestive heart failure and other cardiovascular diseases.
  • SR-202 which not only inhibits hormone and TZD-induced adipocyte differentiation in vitro, but also inhibits adipocyte hypertrophy and insulin resistance in a high-fat-induced obese mouse model
  • Rieusset J et al. A new selective Peroxisome proliferator-activated receptor gamma antagonist with antiobesity and antidiabetic activity.
  • a selective peroxisome proliferator-activated receptor gamma antagonist with anti-diabetic activity against the moon and the moon Mol Endocrinol 16, 2002: 2628-2644
  • In vivo experiments with GW9662 and SR-202 and mice with loss of PPAR Y function were obtained. Similar results, suggesting that reducing the activity of PPAR Y does not cause glucose intolerance. Taken together, these experimental data suggest that PPAR Y antagonists may become the treatment of obesity drugs.
  • Tanshinone is isolated from the lipotype plant Monochamus sinensis 03 ⁇ 4/ a miltiorrhiza Bunge) and is widely used in traditional Chinese medicine for coronary heart disease, cerebrovascular and cardiovascular diseases. Due to its bioactivity and clinically reported side effects, many traditionally important preparations containing single ginseng have been developed.
  • Tanshinone oxime is a lipophilic biguanide compound and is a rich active ingredient in tanshinone. Purified tanshinone sulfonate is effective in stroke heart attacks and angina pectoris, and is effective in reducing brain damage volume in a mouse transient local cerebral ischemia model. It has been reported that the therapeutic effect of tanshinone oxime on atherosclerosis is caused by its antioxidant and anti-inflammatory properties.
  • the present invention provides an application of tanshinone oxime in the preparation of a medicament for the treatment of obesity and insulin resistance, which can be used as a natural medicine which does not promote side effects of fat formation and cardiovascular dysfunction.
  • tanshinone oxime a physiologically or pharmaceutically acceptable salt thereof, a hydrate thereof or a mixture thereof for the preparation or prevention of a disease associated with obesity and/or diabetes Substance.
  • the substance is for reducing body weight and/or blood lipids in an obese patient.
  • the substance is used to reduce the body weight of a mammal, Lower blood lipid levels and / or used to relieve insulin resistance.
  • the obesity-related disease or condition is selected from the group consisting of obesity, diabetes, atherosclerosis, and/or hyperlipidemia.
  • the diabetes-related disease or condition is selected from the group consisting of insulin resistance, hyperglycemia, type II diabetes, hyperinsulinemia, diabetic ketoacidosis, hyperosmolar nonketosis Diabetic coma, and / or lactic acidosis.
  • the tanshinone oxime is extracted or purified from natural plants.
  • the composition contains purified tanshinone oxime sulfonate or hydroxy tanshinone
  • the substance is a composition.
  • the composition is selected from the group consisting of: a pharmaceutical composition, a nutraceutical composition, a food composition or a dietary supplement.
  • the tanshinone oxime accounts for the total weight of the composition.
  • tanshinone oxime comprises from 1 to 95% by weight, more preferably from 5 to 90% by weight, still more preferably from 10 to 80% by weight, based on the total weight of the composition.
  • the substance is in a unit dosage form or a multi-dose form, and wherein the content of tanshinone oxime is from 1 to 3500 mg per dose.
  • the content of tanshinone oxime in the substance is from 1 to 10 mg/dose, more preferably from 2 to 8 mg/dose, further preferably from 3 to 6 mg/dose.
  • the content of tanshinone oxime in the substance is 10-1000 mg / dose; more preferably 20 - 800 mg / dose, further preferably 50 - 600 mg / dose, more preferably 75 - 500 mg / dose.
  • the content of tanshinone oxime in the substance is from 1000 to 3500 mg / eq; more preferably from 1100 to 3200 mg / dose, further preferably from 1200 to 3000 mg / dose, more preferably from 1500 to 2000 mg / dose.
  • the daily application amount is usually 0.01-100 mg tanshinone IIA/kg body weight, preferably 0.05-80 mg tanshinone IIA/kg body weight, more preferably 0.1-60 mg tanshinone IIA/kg body weight, more preferably 0.5-50 mg tanshinone IIA/kg body weight, more preferably 1-25 mg tanshinone IIA/kg body weight, most preferably 2-20 mg tanshinone IIA/kg body weight.
  • composition further comprises weight loss, prevention and
  • / or other substances for the treatment of diabetes more preferably other substances selected from one or more selected from the group consisting of: tolylamine, benzylpropylamine, o-chloroaprofen, benzomorpholine, cyclomethine, Chlorpheniramine, fenfluramine, fluoxetine, AD9677, CP-33 U 679, western komin, orlistat; metformin, phenformin, glibenclamide, glipizide, glibenclamide Qi, glibenclamide, glimepiride, gliclazide, Acarbose, voglibose, miglitol, ciglitazone, troglitazone, rosiglitazone, pioglitazone, astatin, epalrestat, polastatin, or Risostat, repaglinide or nateglinide; or a mixture thereof.
  • a method of weight loss for non-therapeutic purposes comprising administering to a subject in need of weight loss an effective amount of tanshinone oxime, a physiologically or pharmaceutically acceptable salt thereof, a hydrate thereof or a mixture of them.
  • a composition comprising:
  • a pharmaceutically acceptable carrier or a food acceptable carrier (2) A pharmaceutically acceptable carrier or a food acceptable carrier.
  • the tanshinone oxime comprises from 0.001 to 99.9% by weight of the total weight of the composition.
  • the content is from 1 to 95% by weight, more preferably from 5 to 90% by weight, still more preferably from 10 to 80% by weight.
  • the weight ratio of the component (1) to the component (2) is 0.01 : 100 100 : 0.01.
  • tanshinone oxime a physiologically or pharmaceutically acceptable salt thereof, a hydrate thereof or a mixture thereof for the preparation of a disease for the treatment of amelioration by antagonizing PPAR Y or Disordered substance.
  • the disease or disorder is overweight, hyperlipidemia and/or insulin resistance.
  • the tanshinone oxime is extracted or purified from natural plants.
  • the composition contains purified tanshinone oxime sulfonate or hydroxy tanshinone oxime.
  • the substance is a composition.
  • the composition is selected from the group consisting of: a pharmaceutical composition, a nutraceutical composition, a food composition, or a dietary additive.
  • the tanshinone oxime comprises from 0.001 to 99.9% by weight, preferably from 1 to 95% by weight, more preferably from 5 to 90% by weight, still more preferably from 10 to 80% by weight, based on the total weight of the composition.
  • Other aspects of the invention will be apparent to those skilled in the art from this disclosure.
  • Tanshinone oxime inhibits adipocyte differentiation and related gene expression
  • A The chemical structure of tanshinone oxime.
  • B Tanshinone oxime inhibits differentiation of adipocytes induced by differentiation fluid. Oil red staining was performed on the fifth day of differentiation.
  • GM 3T3-L1 cell control;
  • DM differentiation-induced 3T3-L1;
  • DM+Ros differentiation of 3T3-L1 induced by addition of 1 ⁇ rosiglitazone;
  • DM+Tan 5, DM+Tan 25 or DM+ Tan 50 The differentiation fluid induces 3T3-L1 differentiation while adding 5, 25, or 50 ⁇ tanshinone oxime.
  • Flow cytometry for PI and Annexin V staining.
  • Con 3T3-L1 culture was added to DMSO control; Tan 5 , Tan 25 or Tan 50 : 3T3-L1 cells were differentiated, and 5 ⁇ , 25 ⁇ and 50 ⁇ tanshinone oxime were added, respectively.
  • Tanshinone ⁇ inhibits the sensitivity of adipocyte differentiation to time.
  • G-H Tanshinone ⁇ inhibits C/EBP a mRNA and protein levels in a time-sensitive manner.
  • 1 3T3-L1 cell control; 2: differentiation-induced 3T3-L1 cells; 3-5: 50 ⁇ tanshinone oxime was added on day 0-2 of adipocyte differentiation.
  • GM 3T3-L1 cell control
  • DM differentiation-induced 3T3-L1
  • DM+Ros differentiation with 1 ⁇ M rosiglitazone induced 3T3-L1 differentiation
  • DM+Ros+Tan 5 DM+Ros+Tan 25
  • DM+Ros+Tan 50 The differentiation solution induces adipocyte differentiation while adding 1 ⁇ of rosiglitazone and 5 ⁇ , 25 ⁇ and 50 ⁇ tanshinone oxime, respectively.
  • Tanshinone oxime can compete with [ 3 H]-labeled rosiglitazone for binding to PPAR Y.
  • Non-labeled rosiglitazone (BRL49653) was used as a positive control.
  • E SPR analysis verified the binding of PPAR Y to tanshinone IIA.
  • A-B Changes in body weight and fat content in mice treated with normal diet and high fat diet with tanshinone oxime or DMSO.
  • D and E Changes in body weight and fat content of obese mice treated with tanshinone oxime or DMSO.
  • C and F Hematoxylin and eosin staining of white fat sections of mice in the prevention and treatment groups.
  • G-H Changes in white adipose tissue and liver gene expression in obese mice treated with tanshinone oxime. Among them, “HF” stands for high-fat meal induction; “NF” stands for normal diet.
  • Tanshinone oxime reduces insulin levels in the prophylactic and therapeutic groups.
  • D-E Mouse liver protein immunoblotting analysis, tanshinone oxime enhances phosphorylation of Akt and AMPK.
  • a and C Serum TCHO, TG, LDL-c and HDL-c levels in the prophylactic and therapeutic groups.
  • B and D Tanshinone oxime reduced the LDL-c/HDL-c ratio in the prophylactic and treatment groups.
  • tanshinone oxime can inhibit adipocyte differentiation and inhibit weight gain. Furthermore, the inventors have also demonstrated that tanshinone oxime is a natural antagonist of PPARY and provides a new concept of tanshinone oxime in adipocyte differentiation and insulin resistance. This study provides an important basis for the development of therapeutic drugs for obesity and metabolic syndrome. On the basis of this, the present invention has been completed.
  • the inventors mainly discuss the therapeutic effect of tanshinone oxime in an obese patient with a background of metabolic syndrome and the mechanism of its action.
  • This study describes the role of tanshinone oxime as a natural antagonist of PPAR y in regulating adipocyte differentiation. Tanshinone oxime also exhibits a wide range of synergistic effects in improving insulin resistance and lipid metabolism. This is the first report of a multiple synergistic effect of a natural compound tanshinone oxime in obesity and related metabolic syndrome.
  • PPAR Y plays an important role in lipid formation. It has been reported that ectopic expression and activation of PPAR Y in fibroblast cells can induce a lipid-forming response. Genomic studies based on chimeric mice have also demonstrated that PPAR Y is also required for lipid formation in vivo. Synthetic PPAR Y antagonists such as BADGE or SR-202 can inhibit adipocyte differentiation in vitro.
  • tanshinone oxime inhibits lipid accumulation in 3T3-L1 cells, and that tanshinone oxime can also inhibit PPAR ⁇ transcriptional activity in 293T cells co-transfected with full-length PPAR ⁇ plasmid and PPRE fluorescent plasmid, so The inventors have reason to speculate that tanshinone oxime is an antagonist of PPAR Y.
  • SPA binding test proves Dan The ketamine IIA is indeed an antagonist with moderate affinity for PPAR Y.
  • the SPR assay also demonstrated that tanshinone oxime binds to the ligand binding domain of PPAR Y like rosiglitazone and exhibits a dose-dependent profile. Tanshinone oxime as a PPAR Y antagonist effectively inhibits the transcriptional activity of PPAR Y and its target gene.
  • Antagonists of PPAR Y have been reported to block high fat-induced obesity and improve insulin sensitivity.
  • tanshinone oxime can also prevent obesity induced by high-fat diets, and it also has therapeutic effects on obese mice.
  • tanshinone oxime not only reduces body weight, improves insulin resistance, but also reduces blood lipid levels.
  • tanshinone II A can reduce the ratio of LDL-c/HDL-c, which is an important marker of heart attack.
  • the improvement in insulin resistance in obese mice treated with tanshinone oxime is of concern.
  • the results of the glucose tolerance test showed that the treatment of tanshinone oxime not only improved high insulinemia but also improved insulin resistance.
  • the Akt and ⁇ pathways are thought to be involved in the insulin signaling pathway.
  • Inventors using the tanshinone oxime-treated mouse liver protein for immunoblotting experiments found that both Akt and AMPK are activated by tanshinone oxime, indicating that the effect of tanshinone oxime on insulin resistance is through these two pathways.
  • the first explanation for this contradiction is the number and size of fat cells.
  • One is a cytokine that is inhibited by PPAR Y agonists and can affect energy balance and insulin sensitivity, such as TNF- ⁇ .
  • TNF- ⁇ cytokine that is inhibited by PPAR Y agonists and can affect energy balance and insulin sensitivity, such as TNF- ⁇ .
  • tanshinone oxime can reduce LPS-induced IL-8 and VCAM-1 levels and inhibit the expression of pro-inflammatory factors such as N0, TNF-a, IL-1 ⁇ and IL-6 in macrophages RAW 264. Since inflammatory factors have an effect on obesity and diabetes, the anti-inflammatory properties of tanshinone oxime may be beneficial to obese individuals.
  • PGC-1a is expressed in gray adipose tissue higher than white fat, which is usually reflected in cold exposure and is considered a strategy to control fat content.
  • Ectopic expression of PGC-1 a in human white fat induces white fat to differentiate into gray fat. This change at the genetic level promotes the oxidation of fatty acids. This suggests that tanshinone oxime may promote the conversion of white fat to gray fat. However, this requires further verification.
  • tanshinone oxime a well-known traditional Chinese medicine for cardiovascular protection
  • tanshinone oxime can effectively inhibit adipocyte differentiation and lipid accumulation, and can also treat type 2 diabetes by multi-molecular target treatment of high-fat meal-induced weight gain and insulin resistance to promote insulin sensitivity.
  • tanshinone oxime active ingredient
  • tanshinone oxime active substance are used interchangeably and refer to tanshinone oxime, a physiologically acceptable salt thereof or a pharmaceutically acceptable salt, derivative, hydration. Or a mixture thereof, including but not limited to: tanshinone oxime, its sulfonate and hydroxy tanshinone oxime.
  • tanshinone oxime there is no particular limitation on the method for preparing tanshinone oxime, for example, it may be extracted from natural plants, or may be prepared by chemical synthesis or semi-chemical synthesis.
  • the role of tanshinone may be extracted from natural plants, or may be prepared by chemical synthesis or semi-chemical synthesis.
  • Tanshinone has been used in the prevention and treatment of various diseases in previous studies, for example, it has been used in the prevention and treatment of diseases such as coronary heart disease, cerebrovascular and cardiovascular diseases. However, there are no reports on its prevention and treatment of obesity and/or diabetes and/or alleviation of insulin resistance.
  • the present inventors have experimentally demonstrated that tanshinone oxime is a natural PPAR y antagonist, and thus it can be used as a PPAR y antagonist to intervene and treat various diseases involving PPAR ⁇ . Its main effects include but are not limited to the following:
  • the inventors In order to study the weight-loss effect of tanshinone oxime, the inventors first induced high-fat foods to induce obese mice and established an obesity model. Obese rats were treated with tanshinone oxime by gavage. After feeding, the weight, the body weight, the physiological and biochemical indexes of the drug-administered group and the high-fat control group were compared to verify the weight-loss effect of Danshen Ketone.
  • the present invention demonstrates that tanshinone oxime has a good weight loss effect. Because it is a naturally occurring substance in foods and animals, it is safe and can be used as a safe and reliable weight loss active substance.
  • tanshinone oxime active substance has a weight loss effect, it can also be used for preventing, treating and controlling diseases related to obesity, such as but not limited to: diabetes, fatty liver, liver hypertrophy, abnormal lipid metabolism, hyperlipidemia, arteries Atherosclerosis, coronary heart disease, cancer, gout, endocrine disorders, alveolar hypoventilation syndrome, cholecystitis, etc.
  • Tanshinone oxime is especially effective in lowering blood lipids, especially in obesity caused by obesity.
  • the present invention provides the use of a tanshinone oxime active substance for the preparation of a pharmaceutical composition, a food composition, a nutraceutical composition and/or a dietary supplement for preventing, controlling and treating obesity.
  • a pharmaceutical composition for the preparation of a pharmaceutical composition, a food composition, a nutraceutical composition and/or a dietary supplement for preventing, controlling and treating obesity.
  • the tanshinone oxime active substance can also be used for weight loss for non-therapeutic purposes.
  • tanshinone oxime has a blood sugar lowering effect and ameliorating insulin resistance in high fat obese mice, and thus can be used for preparing a medicine or a health care product for preventing and treating diabetes.
  • mice administered tanshinone IIA were studied.
  • the present invention demonstrates that tanshinone oxime is effective in relieving insulin resistance and lowering blood sugar. Because it is a natural substance, it is safe and can be used as a safe and reliable active substance for relieving insulin resistance and lowering blood sugar, thus being applied to the prevention and treatment of diabetes.
  • tanshinone oxime since tanshinone oxime has an effect of alleviating insulin resistance, it can also be used for preventing, treating, and controlling diseases associated with decreased insulin sensitivity, such as, but not limited to, type 2 diabetes, hyperinsulinemia. It can also be used for other related diseases and symptoms caused by insulin resistance, such as: diabetic ketoacidosis, hyperosmotic non-ketotic diabetic coma, lactic acidosis.
  • the present invention provides a use of a tanshinone oxime active substance in the preparation of a pharmaceutical composition, a food composition, a nutraceutical composition and/or a dietary supplement for preventing, controlling and treating diseases or symptoms of diabetes.
  • a pharmaceutical composition a food composition, a nutraceutical composition and/or a dietary supplement for preventing, controlling and treating diseases or symptoms of diabetes.
  • the present invention also provides a novel use of a commercially available tanshinone oxime preparation for controlling blood sugar, relieving insulin resistance, and thereby preventing diabetes.
  • the tanshinone oxime active substance of the present invention can be used for the simultaneous production of a dual effect of preventing, controlling and treating diabetes and treating obesity.
  • the invention also provides a composition and its use in controlling body weight, blood glucose and/or insulin resistance, said composition comprising an effective amount of tanshinone oxime, and a pharmaceutically or pharmaceutically acceptable carrier.
  • the term "pharmaceutically acceptable” or “food acceptable” ingredients are suitable for use in humans and/or animals without excessive adverse side effects (eg, toxicity, irritation, and allergies), ie, A substance with a reasonable benefit/risk ratio.
  • composition of the invention includes pharmaceutical compositions, food compositions, health care compositions, and/or dietary supplements.
  • the term "effective amount” refers to an amount that is functional or active to a human and/or animal and that is acceptable to humans and/or animals.
  • the term "pharmaceutically acceptable carrier” refers to a carrier for the administration of a therapeutic agent, including various excipients and diluents.
  • the term refers to pharmaceutical carriers which are not themselves essential active ingredients and which are not excessively toxic after administration. Suitable carriers are well known to those of ordinary skill in the art. In Remington's Pharmaceutical Sciences, Mack Pub. A full discussion of pharmaceutically acceptable excipients can be found in Co., NJ 1991).
  • composition of the present invention can also be administered as a food or dietary supplement directly or added to other foods.
  • the "food-acceptable carrier” is selected from the group consisting of: a filler, a disintegrant, a lubricant, a glidant, an effervescent agent, a flavoring agent, a coating material, a dietary product, an excipient, A slow/controlled release agent.
  • the pharmaceutically acceptable carrier in the composition may contain a liquid such as water, saline, glycerol and ethanol.
  • auxiliary substances such as fillers, disintegrants, lubricants, glidants, effervescent agents, wetting or emulsifying agents, flavoring agents, pH buffering substances and the like may also be present in these carriers.
  • these materials can be formulated in a non-toxic, inert, and pharmaceutically acceptable aqueous carrier medium wherein the pH is usually from about 5 to about 8, preferably, the pH is from about 6 to about 8.
  • composition of the present invention may contain a physiologically acceptable salt or a pharmaceutically acceptable salt, a hydrate or a mixture thereof of tanshinone oxime, tanshinone oxime.
  • a person skilled in the art can determine the dosage of tanshinone oxime as needed.
  • the daily application rate of the tanshinone oxime composition of the present invention is usually 0.01-100 mg tanshinone IIA/kg body weight, preferably 0.05-80 mg tanshinone IIA/kg body weight, more preferably 0.1.
  • tanshinone IIA/kg body weight more preferably 0.5-50 mg tanshinone IIA/kg body weight, more preferably 1-25 mg tanshinone IIA/kg body weight, most preferably 2-20 mg tanshinone IIA/kg body weight.
  • the active ingredient of the tanshinone oxime active ingredient in the composition of the present invention is from 0.001 to 99.9% by weight based on the total weight of the composition; preferably from 1 to 95% by weight, more preferably from 5 to 90% by weight, still more preferably from 10 to 80% by weight based on the total weight of the composition. .
  • the balance is a pharmaceutically acceptable carrier and other additives.
  • unit dosage form refers to a dosage form required to prepare a composition of the present invention for single administration for ease of administration, including but not limited to various solid agents (eg, tablets;), liquid agents, capsules. Agent, slow release agent.
  • the composition is in a unit dosage form or a multi-dosage form, and the content of the tanshinone oxime active substance is from 1 to 3500 mg per dose.
  • the composition is in unit dosage form or in multiple dosage forms, and wherein the content of tanshinone oxime is from 1 to 3500 mg per dose.
  • the content of tanshinone oxime in the composition is from 1 to 10 mg/dose, more preferably from 2 to 8 mg/dose, still more preferably from 3 to 6 mg/dose.
  • the content of tanshinone oxime in the composition is 10-1000 mg / dose; more preferably 20-800 mg / dose, further preferably 50 - 600 mg / dose, more preferably 75 - 500 mg / dose.
  • the content of tanshinone oxime in the composition is from 1000 to 3500 mg/dose; more preferably from 1100 to 3200 mg/dose, further preferably from 1200 to 3000 mg/dose, more preferably from 1500 to 2000 mg/dose.
  • 1-6 doses of the composition of the invention are administered per day, preferably 1-3 doses; at high doses, preferably 1 dose per day.
  • the effective dose of the tanshinone oxime active substance used may vary with the subject to be administered or treated. Change in degree. The specific situation is determined according to the individual condition of the subject (for example, the subject's weight, age, physical condition, desired effect); this is within the range that the skilled physician or dietitian can judge.
  • composition of the present invention may be in the form of a solid (e.g., granules, tablets, lyophilized powder, suppository, capsule, sublingual tablet; or liquid (e.g., oral solution;) or other suitable shape.
  • a solid e.g., granules, tablets, lyophilized powder, suppository, capsule, sublingual tablet
  • liquid e.g., oral solution;
  • compositions of the present invention can be administered by conventional routes including, but not limited to,: oral, intramuscular, subcutaneous, and the like. Oral administration is preferred.
  • the form of the composition should match the mode of administration.
  • the amount of the composition of the present invention is usually from 0.01 to 100 mg per day of tanshinone IIA / kg body weight, preferably from about 0.05 to 80 mg of tanshinone IIA / kg body weight, preferably 0. 1-60 mg tanshinone IIA /
  • the kg body weight is more preferably 0.5-50 mg/kg body weight, more preferably 1-25 mg tanshinone IIA/kg body weight, and most preferably 2-20 mg tanshinone IIA/kg body weight.
  • compositions of the invention may be used as such or in combination with other therapeutic agents or adjuvants.
  • the composition of the present invention may also be combined with an effective amount (for example, 0.0005-0. 1 g/kg body weight / when used for weight loss or prevention, control, treatment, and obesity-related diseases. Days; preferably, from 0.001 to 0.05 g/kg body weight/day;) a combination of substances selected from the group consisting of: amphetamine and its derivatives, including but not limited to: tolylamine, benzyltolylamine, o-chloro Amphetamine, morpholine; steroids, including but not limited to cyclomethine, chlorpheniramine; serotoninergic drugs, including (but not limited to): fenfluramine, fluoxetine e 3 -AR agonists, including but not limited to: AD9677, CP-33 U 679; monoamine reuptake inhibitors, including (but not limited to): western koji; lipase inhibitors, including (but Not limited to ;): Orlistat and so
  • an effective amount for example,
  • the composition of the present invention may also be administered in an amount effective to prevent, treat, and alleviate diabetes, particularly insulin resistance, (e.g., 0.5 to 100 mg/kg body weight/day; preferably a combination of substances selected from the group consisting of l-50 mg/kg body weight/day;):
  • the drugs of the biguanide include, but are not limited to: metformin, or phenformin; sulfonylurea diabetes drugs include ( But not limited to ;): glibenclamide, glipizide, gliclazide, glibenclamide, glimepiride, or gliclazone; glucosidase inhibitors include (but are not limited to Acarbose, voglibose (; vokibose;), or miglitol; insulin sensitizing drugs including (but not limited to;): ciglitazone, troglitazone, rosiglitazone, Or piogli
  • insulin resistance e.
  • the co-administered drug or other formulation does not interfere with the therapeutic activity of the active ingredient of the tanshinone oxime active of the present invention.
  • the invention also provides a method of weight loss for non-therapeutic purposes, the method comprising administering to the subject an effective amount of a tanshinone oxime active.
  • the amount of the tanshinone oxime active substance administered to the subject is: usually about 0.001 to 100 mg of tanshinone IIA / kg body weight per day, preferably 0.01 to 100 mg of tanshinone IIA / kg body weight, preferably about 0.05. - 80 mg tanshinone IIA/kg body weight, preferably 0.1-60 mg tanshinone IIA/kg body weight, more preferably 0.5-50 mg/kg body weight, more preferably 1-25 mg tanshinone IIA/kg body weight, most preferably 2-20 mg tanshinone IIA /kg body weight.
  • the amount may be administered in a single administration or multiple times during the day.
  • the weight loss method further comprises administering to the subject other weight loss drugs or health care products, such as those described above.
  • the weight loss method further includes other weight loss regimens or measures, such as exercise, dieting, and the like.
  • the present invention discloses a novel use of tanshinone oxime in the treatment of obesity, blood sugar and insulin sensitivity, and thereby develops a novel health care product, medicine or dietary additive containing tanshinone oxime.
  • the present invention has the following advantages:
  • the tanshinone oxime used in the present invention is a naturally occurring substance in foods and animals, and has been mainly used for food additives and synthesis in previous studies. Drugs, etc., have high safety;
  • tanshinone oxime has been used in the prevention and treatment of various diseases, such as coronary heart disease, cerebrovascular and cardiovascular diseases, and is therefore easily accepted by the market;
  • tanshinone oxime has the characteristics of good stability and small side effects, and is more suitable for development as a new type of weight loss health care product or medicine.
  • Tanshinone oxime inhibits adipocyte differentiation and inhibits gene differentiation of 3T3-L1 cells
  • Tanshinone IIA (Tanshinone IIA, Tan)
  • Mouse preadipocytes 3T3-L1 were purchased from ATCC, medium DMEM plus 10% fetal bovine serum (growth medium).
  • growth medium 10% fetal bovine serum
  • the cells are transferred to a 12-well or 24-well plate, and the cells are cultured for 2 days after the cells are fully confluent, and then dexamethasone containing 10 g/ml of insulin, 50 ⁇ M, and 0.8 mM of sputum.
  • the growth medium (differentiation fluid) induces differentiation. After 4 days of differentiation, the differentiation medium was changed to DMEM plus 10% FBS growth medium. At the time of differentiation, the cells were cultured at 37 ° C in 10% C0 2 conditions.
  • Cell processing (The cell processing conditions described herein include cell processing conditions throughout the assay)
  • the differentiation medium When the differentiation medium was used to induce adipocyte differentiation, 1 ⁇ of rosiglitazone and/or 5, 25, 50 ⁇ M of tanshinone oxime was added to the differentiation solution.
  • 1 ⁇ of rosiglitazone was added to the differentiation solution while differentiation, and on the 0th day of differentiation (ie, simultaneous differentiation;), day 1, day 2, day 3 and On day 4, 50 ⁇ M of tanshinone oxime was added to the differentiation solution.
  • the 3T3-L1 cells were fixed with 10% formalin for 10 minutes at room temperature, the cells were washed twice with PBS, stained with oil red at 60 ° C for 15 minutes, and washed twice with PBS, followed by an Olympus microscope (1X71). Take a photo.
  • Tissue and cellular total RNA was extracted using the Qiagen RNA kit.
  • Single-strand cDNA was synthesized using a promega cDNA synthesis kit.
  • Gene expression levels were analyzed using an ABI 7500 real-time quantitative PCR system.
  • the cDNA was kept at 50 ° C for 2 minutes and amplified for 40 cycles (95 ° C, 15 s, 60 ° C, 60 s) after denaturation at 95 ° C for 10 minutes. All trials were repeated at least 3 times.
  • Gene mRNA levels were made with ⁇ -actin ( ⁇ -actin) as an internal reference.
  • Tissues and cells were directly lysed with 2XSDS loading buffer and boiled for 5 minutes.
  • the 10 total protein was electrophoresed on a 10% SDS-PAGE gel and transferred to a PVDF membrane.
  • the transferred membrane was blocked with a 5 % skim milk powder solution in a TBS-T buffer for 1 hour, and then the antibody diluted with 1% BSA was added at 4 ° C overnight.
  • the antibody-coated membrane was washed 3 times with TBS-T buffer for 10 minutes each time, and HRP-labeled secondary antibody was added to the room for 1 hour, then washed with TBS-T for 3 hours, and finally developed with ECL coloring solution.
  • annexin V-FITC/PI apoptosis assay kit (Si gma). 3T3-L1 cells were harvested and stained with annexin V-FITC/PI and analyzed with a BD flow meter (BD FACSAr ia).
  • test results and analysis were analyzed by SPSS 12. 0, and statistical analysis was performed by one-way analysis of variance (AN0VA). Statistical significance was considered when the P value was less than 0.05. Test results and analysis
  • tanshinone oxime inhibits the differentiation of fat cells.
  • Flow cytometry analysis showed that the treatment of tanshinone oxime did not increase the number of cells that were apoptotic and dead, indicating that the inhibition of adipocyte differentiation gene expression by tanshinone oxime was not caused by apoptosis or toxicity (Fig. 1E).
  • the inhibitory effect of tanshinone oxime on adipocytes is the strongest in the treatment of differentiation.
  • the results of oil red staining showed that the inhibitory effect of the addition of tanshinone oxime on the second day or later of differentiation was attenuated, and the inhibitory effect of tanshinone oxime on the RNA and protein levels of C/EBP ci was also weakened (Fig. 1F, G and H).
  • the Gal4-PPAR Y ligand binding domain plasmid is co-transferred into the cells with the UASG X 4-TK-Luc reporter plasmid and the jellyfish fluorescent plasmid, and the dosing and analysis methods are the same as above.
  • SPA analysis was performed to test the affinity of PPAR Y for binding to tanshinone oxime. Briefly, biotin-labeled PPRE was mixed with a hair buffer containing fish sperm DNA, 4 mg of streptavidin-coated beads, and placed in a centrifuge tube at 4 ° C overnight. The mixture was centrifuged at 1500 x g for 10 minutes. Wash 3 times with 10 ml of reaction buffer after the supernatant.
  • the Biacore instrument is designed based on the principle that P-polarized light penetrates into the metal film when total internal reflection occurs at the interface between the glass and the metal film, and the free electrons in the metal generate surface plasmons. Biosensors enable unlabeled bioreactive signals to be delivered to the monitor in real time. Binding assay using BIAcore TM 3000 is completed at the instrument, 25 ° C. ⁇ ⁇ protein is immobilized on the surface of the chip. Tanshinone oxime is injected into the surface of the chip as a solution. When tanshinone oxime reacts with ⁇ immobilized on the surface of the chip, changes in the SPR response are detected. Changes in the level of response are recorded in equal proportions with changes in the module.
  • tanshinone oxime inhibits gene expression in the PPAR Y pathway, it is speculated that tanshinone oxime inhibits adipocyte differentiation by directly regulating PPAR Y activity.
  • a PPAR Y expression plasmid and a corresponding PPAR Y signal PPRE fluorescent reporter gene were co-transfected into 293 cells. It was first verified that tanshinone oxime inhibits the transcriptional activity of full-length PPAR ⁇ in the presence and absence of rosiglitazone (Fig. 2 ⁇ ). Furthermore, the same inhibitory effect was observed in the GAL4-PPAR ⁇ LBD fusion plasmid system which excluded the intrinsic ligand interference (Fig. 2 ⁇ ).
  • tanshinone oxime is a ligand for PPAR gamma
  • a competitive binding assay was designed to simultaneously add a strong agonist of PPARy, rosiglitazone (1 ⁇ ) and tanshinone oxime, in the process of adipocyte differentiation. Staining showed that tanshinone oxime was still effective in inhibiting adipocyte differentiation (Fig. 2C).
  • has a moderate affinity with PPARy (Fig. 2D).
  • the SPR assay verified the direct binding of tanshinone oxime to PPARy (KD, 6. 74 x 10- 5 M) (Fig. 2E).
  • Example 3 Therapeutic effect of tanshinone oxime on obese mice induced by high fat diet
  • mice Female C57BL/6J mice were purchased from SLAC Laboratories. All animals were housed at 22 - 23 ° C for 12 hours and 12 nights. C57BL/6J mice were fed with a 60% fat content feed to induce an obesity model, and a 10% fat content feed-fed mouse was used as a control. Mice were intragastrically administered with 35 mg of tanshinone IIA/kg body weight (200 ⁇ l) or 200 ⁇ l of DMS0 at 1 pm every other day. Serum triglycerides, total cholesterol, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol were analyzed using a Hi tachi 7020 automated analyzer.
  • White adipose tissue was embedded in OCT, sliced into 10- ⁇ ⁇ sections by frozen sectioning, and then stained with hematoxylin and eosin. Test results and analysis
  • tanshinone oxime is an antagonist of PPAR Y and can inhibit adipocyte differentiation in vitro
  • the role of tanshinone oxime in the fat content and body weight change of high-fat diet-induced and normal-fed C57BL/6 mice was examined.
  • tanshinone oxime treatment was effective in preventing weight gain induced by high fat meal compared with the control group.
  • the fat cells in the white fat tissue of the high fat-induced mouse in the tanshinone oxime treatment group were smaller than those in the control group (Fig. 3C). There was no significant difference in the body weight of the tanshinone oxime treatment group and the control group in the normal feed-fed mice.
  • mice C57BL/6J mice were induced with high fat for 5 months and their body weight increased to 150%. Obese mice were divided into two groups and treated with tanshinone oxime (35 mg/kg) or DMS0 (control) every other day for 9 weeks. The results showed that the mice in the tanshinone oxime treatment group had significantly lower body weight than the control group, and the former had a smaller fat cell volume than the control group (Fig. 3D, E and F).
  • IPGTT Intraperitoneal glucose tolerance test
  • High-fat meal-induced C57BL/6J mice were treated with tanshinone oxime or DMS0 (control) for 2 months and fasted for 8 hours, intraperitoneal injection of 2 g/kg body weight of glucose, 0, 15, 30, 60 when glucose was injected. Blood glucose was measured from the tail vein in 120 minutes (0 minutes is the fasting blood glucose when no glucose is injected). Immunoblotting
  • Tissues and cells were directly lysed with 2XSDS loading buffer and boiled for 5 minutes.
  • the 10 total protein was electrophoresed on a 10% SDS-PAGE gel and transferred to a PVDF membrane.
  • the transferred membrane was blocked with a 5 % skim milk powder solution in a TBS-T buffer for 1 hour, and then the antibody diluted with 1% BSA was added at 4 ° C overnight.
  • the antibody-coated membrane was washed 3 times with TBS-T buffer for 10 minutes each time, and HRP-labeled secondary antibody was added to the room for 1 hour, then washed with TBS-T for 3 hours, and finally developed with ECL coloring solution.
  • Figs. 4A and 4B the serum insulin level of the control mice induced by the high fat meal was remarkably increased, and the treatment of the tanshinone oxime effectively lowered the serum insulin level. Obesity often causes insulin resistance, which is considered a risk factor for type 2 diabetes, so it is tested whether tanshinone treatment can improve insulin resistance caused by obesity.
  • mice liver tissue protein was extracted for immunoblotting test to detect gene expression related to insulin resistance. The results showed that tanshinone oxime promoted phosphorylation of Akt 308 threonine and 473 serine, and also promoted AMPK.
  • Serum biochemical analysis showed that tanshinone oxime treatment in the prophylactic test group can reduce low-density lipoprotein induced by high-fat diet and normal diet and high-fat-induced triglyceride levels, while high-density lipoprotein in normal diet mice is significantly improved. (Fig. 5A).
  • tanshinone oxime treatment in the treatment test group can reduce the increase in triglyceride high-density lipoprotein and low-density lipoprotein induced by high-fat meal induction (Fig. 5C).
  • the ratio of LDL-c/HDL-c was reduced in all test groups (Fig. 5B and D).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Diabetes (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Emergency Medicine (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Biotechnology (AREA)
  • Botany (AREA)
  • Medical Informatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

丹参酮 ΠΑ在制备治疗肥胖及胰岛素抵抗的药物中的应用 技术领域
本发明涉及生物技术和医药领域, 具体涉及丹参酮 ΠΑ在预防、 控制或治 疗肥胖、 胰岛素抵抗以及糖尿病中的用途。 背景技术
诸多资料报导了越来越多的肥胖人群会发展成为血脂障碍、 高血压、 冠心 病及中风等疾病, 脂肪组织过多通常伴随着胰岛素抵抗和 2型糖尿病风险显著 增加。
过氧化物酶体增殖物激活受体 Y (PPAR γ;)配体激活类核受体超家族的一 个成员, 是一个控制脂肪细胞分化、 脂质形成和葡萄糖代谢等过程基因表达的 主要转录因子。 PPAR γ通过与黄醇类 X受体 (; RXR)形成异二聚体, 并直接结合 到其靶基因的 PPAR反映元件 (PPRE)上而发挥作用的。
获得和缺失功能试验清楚的阐明了 PPAR γ在脂肪细胞发育过程中发挥了 重要的作用, 因此使得 PPAR γ在过去的几十年中成为代谢性疾病相关的药物 靶点。 例如, 噻唑烷二酮类抗糖尿病药物 (TZDs;)、 吡格列酮、 罗格列酮就是已 知的 ΡΡΑΙ γ的激动剂, 它们的胰岛素增敏效果是通过它们在脂肪组织中的多效 性介导的。 PPARj的激动剂, 如 TZDs是当前治疗 2型糖尿病的一线类药物。 然 而, 这些药物具有很多副作用, 包括体重增加、 积水、 肺水肿和黄斑水肿等, 这些又增加了充血性心力衰竭及其它心血管疾病的发病风险。
因此, 研究发展新的以 ΡΡΑΙ γ为靶点、 但没有促进脂肪形成和心血管功能 异常的副作用的更为安全的药物是非常受到关注和重视的。
有研究报导 ΡΡΑΙ γ缺失的杂合子和 ΡΡΑΙ γ的激活都能促进胰岛素抵抗并预 防月旨肪细胞月巴大化 (Yamauchi T等, The mechanisms by which both heterozygous peroxisome proliferator-activated receptor gamma (PPAR gamma) deficiency and PPARgamma agonist improve insulin resistance. "杂合的过氧化物酶体增生物激 活受体 γ(ΡΡΑΙ γ)缺失与 ΡΡΑΙ γ促效剂对胰岛素抵抗改善的共同机理" J Biol Chem 276, 2001 :41245-41254)。 这表明适当的降低 ΡΡΑΙ γ活性可能降低 2型糖 尿病发展的风险。 然而, 对于有代谢性疾病的糖尿病人还没有一种更安全的治 疗药物。 新的研究已经揭示出 ΡΡΑΙ γ拮抗剂的潜在用处, 它们可能既能发挥治 疗作用又能降低副作用。
Wright等报导了一种人工合成的 ΡΡΑΙ γ拮抗剂, 它可以抑制脂肪细胞分化 (Wright HM等, A synthetic antagonist for the peroxisome proliferator-activated receptor gamma inhibits adipocyte differentiation. "过氧化物酶体增生物激活受 体 γ合成拮抗剂对脂肪细胞分化的抑制作用 " J Biol Chem 275, 2000: 1873- 1877)。 GW9662被报导可以预防高脂餐诱导的肥胖 (Nakano R等, Antagonism of peroxisome proliferator-activated receptor gamma prevents high-fat diet-induced obesity in vivo. "过氧化物酶体增生物激活受体 γ拮抗作用在体内防 止了高脂饮食诱导的肥胖症" Biochem Pharmacol 72, 2006:42-52。 另一个例子 是 SR-202 , 它不仅可以在体外抑制激素和 TZD诱导的脂肪细胞分化, 而且在高 脂餐诱导的肥胖小鼠模型中还可以抑制脂肪细胞的肥大化及胰岛素抵抗 (Rieusset J等, A new selective peroxisome proliferator-activated receptor gamma antagonist with antiobesity and antidiabetic activity. "一禾中新的具有抗月巴月半症禾口抗 糖尿病活性的选择性过氧化物酶体增生物激活受体 γ拮抗剂" Mol Endocrinol 16, 2002:2628-2644)。 GW9662和 SR-202的体内试验与 PPAR Y功能缺失的小鼠 得到了相似的结果, 这表明降低 PPAR Y活性不会导致葡萄糖耐受不良。 综上 所述, 这些试验数据说明 PPAR Y拮抗剂有可能成为治疗肥胖的药物。
丹参酮是从唇型科植物单参 0¾/ a miltiorrhiza Bunge)中分离得到的,在传 统的中药中被广泛应用于冠心病, 脑血管和心血管疾病中。 由于它所具备的生 物活性及临床上报道的没有副作用的特性, 研制了很多的含有单参的传统重要 制剂。
丹参酮 ΠΑ是一种亲脂的双萜类化合物, 是丹参酮中含量丰富的活性成分。 纯化的丹参酮 ΠΑ的磺酸盐在中风心脏病发作和心绞痛中是有效的,并且在小鼠 瞬时局部脑缺血模型中可以有效减少脑损伤体积。有报导指出丹参酮 ΠΑ对动脉 粥样硬化的治疗效果是由它的抗氧化和抗炎属性引起的。
然而, 现有技术中并未对丹参酮 ΠΑ与 PPAR Y的关系进行研究, 更没有揭 示丹参酮 ΠΑ对肥胖、 血脂过高或胰岛素抵抗的治疗和缓解作用。
本领域迫切需要开发出一种没有促进脂肪形成和使心血管功能异常的副 作用的更为安全的天然药物。 发明内容
本发明正是提供了丹参酮 ΠΑ在制备治疗肥胖及胰岛素抵抗的药物中的应 用, 它可作为没有促进脂肪形成和心血管功能异常的副作用的天然药物使用。 在本发明的第一方面, 提供了丹参酮 ΠΑ、 其生理学上或药学上可接受的盐、 其水合物或它们的混合物的用途, 其用于制备预防或治疗与肥胖和 /或糖尿病相关 的疾病的物质。
在本发明的一个优选例中, 所述物质用于降低肥胖症患者的体重和 /或血脂。 在本发明的一个优选实施方式中, 所述物质用于降低哺乳动物的体重、用于 降低血脂水平和 /或用于缓解胰岛素抵抗。
在另一优选例中, 所述与肥胖相关的疾病或征状选自下组: 肥胖症、糖尿病、 动脉粥样硬化、 和 /或高血脂。
在另一优选例中, 所述与糖尿病相关的疾病或征状选自下组: 胰岛素抵抗、 血糖过高、 II型糖尿病、 高胰岛素血症、 糖尿病酮症酸中毒、 高渗性非酮症糖尿病 昏迷、 和 /或乳酸性酸中毒。
在本发明的另一个优选实施方式中, 所述丹参酮 ΠΑ是从天然植物提取纯化或 人工合成的。
在一个优选例中, 所述组合物中含有纯化的丹参酮 ΠΑ磺酸盐或羟基丹参酮
II A。
在本发明的另一个优选实施方式中, 所述物质是组合物。
在一个优选例中, 所述组合物选自: 药物组合物、 保健品组合物、 食物组 合物或膳食添加剂。
在本发明的另一个优选实施方式中, 所述丹参酮 ΠΑ占所述组合物总重量的
0.001-99.9wto/o
在本发明的一个优选例中, 丹参酮 ΠΑ占组合物总重量的 l-95wt%, 较优选为 5-90wt%, 更优选 10-80wt%。
在本发明的另一个优选实施方式中, 所述物质为单位剂型或多剂型, 且其中 丹参酮 ΠΑ的含量为 l-3500mg/剂。
在本发明的一个优选例中, 所述物质中丹参酮 ΠΑ的含量为 l-10mg/剂, 更优 选为 2-8mg/剂, 进一步优选含有 3-6mg/剂。
在本发明的另一个优选例中, 所述物质中丹参酮 ΠΑ的含量为 10-1000mg/ 剂; 更优选 20-800mg/剂, 进一步优选 50-600mg/剂, 更优选 75-500mg/剂。
在本发明的另一个优选例中, 所述物质中丹参酮 ΠΑ的含量为 1000-3500mg/ 齐 IJ ; 更优选 1100-3200mg/剂, 进一步优选 1200-3000mg/剂, 更优选 1500-2000mg/ 剂。
在本发明的另一个优选例中, 每天施用量通常为 0.01-100 mg 丹参酮 IIA/kg体重, 优选 0.05-80 mg 丹参酮 IIA/kg体重, 更优选 0.1-60 mg 丹参酮 IIA/kg体重,更优选 0.5-50 mg 丹参酮 IIA/kg体重,更优选 1-25 mg 丹参酮 IIA/kg 体重, 最优选 2-20 mg 丹参酮 IIA/kg体重。
在本发明的另一个优选实施方式中,所述组合物中还包括用于减肥、预防和
/或治疗糖尿病的其它物质, 更佳地所述的其它物质为选自下组中的一种或多 种: 甲苯丙胺、 苄甲苯丙胺、 邻氯苯丙胺、 苯甲吗啉、 环咪吲哚、 氯苯咪吲哚、 芬氟拉明、 氟西丁、 AD9677、 CP-33 U 679、 西部曲明、 奧利司他; 二甲双胍、 苯乙双胍、 格列本脲、 格列吡嗪、 格列齐持、 格列波脲、 格列美脲、 格列喹酮、 阿卡波糖、 伏格利波糖、 米格列醇、 环格列酮、 曲格列酮、 罗格列酮、 吡格列 酮、 阿司他丁、 依帕司他、 波拉司他、 或托瑞司他、 瑞格列奈或那格列奈; 或 它们的混合物。 在本发明的第二方面中, 提供了一种非治疗目的的减肥方法, 所述方法包 括给予需要减肥的对象有效量的丹参酮 ΠΑ、 其生理学上或药学上可接受的盐、 其水合物或它们的混合物。 在本发明的第三方面中, 提供了一种组合物, 所述组合物含有:
(1) 有效量的丹参酮 ΠΑ、 其生理学上或药学上可接受的盐、 其水合物或它们 的混合物;
(2) 选自下组的一种或多种物质: 甲苯丙胺、 苄甲苯丙胺、 邻氯苯丙胺、 苯 甲吗啉、 环咪吲哚、 氯苯咪吲哚、 芬氟拉明、 氟西丁、 AD9677、 CP-33 U 679、 西部曲明、 奧利司他; 二甲双胍、 苯乙双胍、 格列本脲、 格列吡嗪、 格列齐持、 格列波脲、 格列美脲、 格列喹酮、 阿卡波糖、 伏格利波糖、 米格列醇、 环格列 酮、 曲格列酮、 罗格列酮、 吡格列酮、 阿司他丁、 依帕司他、 波拉司他、 或托 瑞司他、 瑞格列奈、 那格列奈; 或它们的混合物; 和
(3) 药学上可接受的载体或食品学上可接受的载体。
在一个优选例中, 所述丹参酮 ΠΑ占组合物总重量的 0.001-99.9wt%。
在本发明的另一优选例中, 所述含量为 l-95wt%, 较优选为 5-90wt%, 更优选 10-80wt
在另一优选例中, 组分 (1)和组分 (2)的重量比为 0.01 : 100 100:0.01。 在本发明的第四方面中,提供了丹参酮 ΠΑ、其生理学上或药学上可接受的盐、 其水合物或它们的混合物的用途,其用于制备治疗通过拮抗 PPAR Y而得到缓解的 疾病或失调的物质。
在一个优选例中, 所述疾病或失调是体重过重、 血脂过高和 /或胰岛素抵抗。 在一个优选例中, 所述丹参酮 ΠΑ是从天然植物提取纯化或人工合成的。 在另一个优选例中, 所述组合物中含有纯化的丹参酮 ΠΑ磺酸盐或羟基丹 参酮 ΠΑ。
在另一个优选例中, 所述物质是组合物。
在另一个优选例中, 所述组合物选自: 药物组合物、 保健品组合物、 食物 组合物或膳食添加剂。
在另一个优选例中, 所述丹参酮 ΠΑ占所述组合物总重量的 0.001-99.9wt%, 优选 l-95wt%, 较优选为 5-90wt%, 更优选 10-80wt%。 本发明的其它方面由于本文的公开内容, 对本领域的技术人员而言是显而 易见的。 附图说明
以下结合附图对本发明进行进一步阐述。 在本发明附图中, " Tan"代表丹 参酮 ΠΑ; " Con" 代表对照; " Ros " 代表罗格列酮。
图 1 : 丹参酮 ΠΑ抑制脂肪细胞分化及相关基因表达
A: 丹参酮 ΠΑ的化学结构。
B : 丹参酮 ΠΑ抑制分化液诱导的脂肪细胞分化。 在分化的第五天进行油红 染色。 GM: 3T3-L1细胞对照; DM: 分化液诱导的 3T3-L1; DM+Ros : 加入 1 μΜ 罗格列酮的分化液诱导 3T3-L1分化; DM+Tan 5、 DM+Tan 25或 DM+Tan 50 : 分 化液诱导 3T3-L1分化同时加入 5、 25、 或 50 μΜ 丹参酮 ΠΑ。
C: 实时定量 PCR结果显示分化第五天时 50 μΜ 丹参酮 ΠΑ 抑制 C/EBP α、 aP2、 CD36、 LPL 和 UCP-2表达。
D: 免疫印记结果, 1 : 3T3-L1细胞对照; 2: 分化液诱导的 3T3-L1 ; 3: 加 入 1 μΜ罗格列酮的分化液诱导 3T3-L1分化; 4-6 : 分化液诱导 3T3-L1分化同时 加入 5 μΜ、 25μΜ 禾口 50 μΜ丹参酮 ΠΑ 。
Ε: 流式细胞检测 PI和膜联蛋白 V染色。 Con: 3T3-L1培养加入 DMSO对照; Tan 5 , Tan 25或 Tan 50 : 3T3-L1细胞分化的同时, 分别加入 5 μΜ、 25 μΜ 禾口 50 μΜ 丹参酮 ΠΑ。
F:丹参酮 ΠΑ抑制脂肪细胞分化对时间的敏感性。其中: DM + Tan 50 d0 (;第 0天) - Tan 50 d4(第 4天;): 在脂肪细胞分化的第 0— 4天加入 50 μΜ 丹参酮 ΠΑ。
G-H: 丹参酮 ΠΑ抑制 C/EBP a mRNA和蛋白水平均呈现时间敏感性。 1 : 3T3-L1细胞对照; 2 : 分化液诱导的 3T3-L1细胞; 3— 5: 在脂肪细胞分化的第 0-2天加入 50 μΜ 丹参酮 ΠΑ。
图 2 : 对丹参酮 ΠΑ是 PPAR Y拮抗剂的证明
Α: 全长 PPAR Y报告基因分析。
B : PPAR Y配体结合结构域 GAL4融合质粒报告基因分析。
C: 分化五天的脂肪细胞油红染色。 GM: 3T3-L1细胞对照; DM: 分化液 诱导的 3T3-L1 ; DM+Ros: 加入 1 μΜ罗格列酮的分化液诱导 3T3-L1分化; DM+Ros+Tan 5、 DM+Ros+Tan 25、 DM+Ros+Tan 50: 分化液诱导脂肪细胞分 化同时, 分别加入 1 μΜ罗格列酮和 5 μΜ、 25 μΜ 和 50 μΜ 丹参酮 ΠΑ。
D: 丹参酮 ΠΑ可以与 [3H]标记的罗格列酮竞争性与 PPAR Y结合。非标记罗 格列酮(BRL49653)用作阳性对照。 E: SPR分析验证 PPAR Y与丹参酮 IIA的结合。
图 3: 丹参酮 ΠΑ抑制高脂餐诱导的肥胖
A-B: 正常饮食和高脂餐诱导的小鼠用丹参酮 ΠΑ或 DMSO处理的体重和 脂肪含量的变化。
D和 E: 肥胖小鼠用丹参酮 ΠΑ或 DMSO处理的体重和脂肪含量的变化。 C和 F: 预防组和治疗组小鼠白色脂肪切片的苏木精和伊红染色。
G-H: 丹参酮 ΠΑ处理的肥胖小鼠白色脂肪组织和肝脏基因表达变化。 其中 "HF" 代表高脂餐诱导; "NF"代表正常饮食。
图 4: 丹参酮 ΠΑ在肥胖小鼠中改善胰岛素抵抗
A-B: 丹参酮 ΠΑ降低预防组和治疗组小鼠的胰岛素水平。
C: 禁食的肥胖小鼠葡萄糖耐受试验。
D-E: 小鼠肝脏蛋白免疫印记分析, 丹参酮 ΠΑ增强 Akt和 AMPK的磷酸化 水平。
图 5: 丹参酮 ΠΑ降低肥胖鼠血脂水平
A和 C: 预防组和治疗组小鼠血清 TCHO、 TG、 LDL-c和 HDL-c水平。 B和 D: 丹参酮 ΠΑ降低预防组和治疗组小鼠 LDL-c/HDL-c比值。 具体实施方式
本发明人通过长期而深入的研究发现丹参酮 ΠΑ能抑制脂肪细胞分化、抑制 体重增加。 并且, 本发明人还证明了丹参酮 ΠΑ是一种 PPARY天然的拮抗剂并 且提供了丹参酮 ΠΑ在脂肪细胞分化及胰岛素抵抗发挥作用的一种新的理念。本 研究为肥胖和代谢性综合症治疗药物的发展提供了重要的依据。 在此基础上, 完成了本发明。
具体而言,在本发明中, 发明人主要论述了丹参酮 ΠΑ在伴有代谢性综合症 背景的肥胖病人中的治疗效果以及它作用的机理。本研究阐述了丹参酮 ΠΑ做为 一个 PPAR y天然拮抗剂在调控脂肪细胞分化中的作用。丹参酮 ΠΑ在改善胰岛素 抵抗和脂质代谢中也体现出了广泛的协同作用。 这是首次报导一个天然化合物 丹参酮 ΠΑ在肥胖和相关的代谢综合症中表现出的多重协同作用。
在功能获得和缺失试验均有证据说明 PPAR Y在脂质形成中起重要作用。 有 报导说明在成纤维细胞细胞中异位表达和激活 PPAR Y可以诱导成脂反应。 基于 嵌合鼠的基因组研究也证明了体内的脂质形成也需要 PPAR Y。 人工合成的 PPAR Y拮抗剂如 BADGE或 SR-202可以在体外抑制脂肪细胞分化。 本发明人通过 体外试验证明了丹参酮 ΠΑ可以抑制 3T3-L1细胞的脂质积累,并且在共转了全长 PPAR γ质粒合 PPRE荧光质粒的 293T细胞中丹参酮 ΠΑ也可以抑制 PPAR γ转录活 性, 所以发明人有理由推测丹参酮 ΠΑ是 PPAR Y的拮抗剂。 SPA结合试验证明丹 参酮 IIA确实是与 PPAR Y有中等亲和力的拮抗剂, SPR试验也证明了丹参酮 ΠΑ可 以同罗格列酮一样与 PPAR Y的配体结合结构域结合, 并呈现剂量依赖的特征。 做为 PPAR Y拮抗剂的丹参酮 ΠΑ有效的抑制了 PPAR Y的转录活性及其靶基因。
据报导, PPAR Y的拮抗剂可以阻止高脂餐诱导的肥胖并且提高胰岛素敏感 性。 做为一个新的 PPAR Y拮抗剂, 丹参酮 ΠΑ同样可以预防高脂餐诱导的肥胖, 而且对于已经肥胖的小鼠丹参酮 ΠΑ同样有治疗效果。在肥胖小鼠模型中丹参酮 ΠΑ不仅可以减轻体重, 改善胰岛素抵抗, 而且可以减少血脂含量。 除此之外丹 参酮 II A可以降低 LDL-c/HDL-c的比值, LDL-c /HDL-c是心脏病发作的一个重要 标志。丹参酮 ΠΑ的这些特征尤其对于高血脂和有心脏病并发症的肥胖个体是有 突出的优势。 2型糖尿病的发病有两个主要的特点: 一是胰岛素抵抗, 二是损 伤胰岛 β细胞的胰岛素分泌。丹参酮 ΠΑ处理的肥胖小鼠胰岛素抵抗的改善是值 得注意的。葡萄糖耐受试验结果显示丹参酮 ΠΑ的处理不仅可以改善高胰岛素血 症还可以改善胰岛素抵抗。 Akt和 ΑΜΡΚ通路被认为参与了胰岛素信号通路。 发 明人利用丹参酮 ΠΑ治疗过的小鼠肝脏蛋白做免疫印记试验发现, Akt和 AMPK都 被丹参酮 ΠΑ激活,这说明丹参酮 ΠΑ对胰岛素抵抗的作用是通过了这两条通路。
发明人的试验结果和别的研究人员的报导揭示了同样的一个有趣的矛盾, 即激动剂激活和通过拮抗剂抑制 PPAR Y活性都可以预防高脂餐诱导的肥胖, 胰 岛素抵抗以及 2型糖尿病。 对于这一矛盾的解释一是脂肪细胞的数量和大小, 一是被 PPAR Y激动剂抑制的可以影响能量平衡和胰岛素敏感性的细胞因子, 如 TNF- α。 有报导指出丹参酮 ΠΑ可以在巨噬细胞 RAW 264. 7中降低 LPS诱导的 IL-8 和 VCAM-1水平并抑制 N0, TNF- a , IL-1 β和 IL-6等促炎症因子的表达。 由于炎 症因子有诱发肥胖和糖尿病的作用,因此丹参酮 ΠΑ的抗炎属性可能对肥胖个体 有好处。
发明人的研究中另一个有趣的发现是丹参酮 ΠΑ处理的小鼠白色脂肪组织 中 PGC-l a的表达被显著提高。 PGC-1 a在灰色脂肪组织中表达高于白色脂肪, 它通常对冷暴露起反映, 并且被认为是控制脂肪含量的策略。 在人白色脂肪中 异位表达 PGC-1 a会诱导白色脂肪向灰色脂肪分化。 基因水平上这种变化会促 进脂肪酸的氧化。这暗示丹参酮 ΠΑ有可能促进白色脂肪向灰色脂肪的转变。然 而这需要进一步验证。
综上所述,本发明人发现并证实了丹参酮 ΠΑ这种被人熟知的心血管保护类 传统中药是一种天然的 PPAR Y拮抗剂。在体外丹参酮 ΠΑ可以有效抑制脂肪细胞 分化和脂质积累, 还可以通过多分子靶点治疗高脂餐诱导的体重增加和胰岛素 抵抗促进胰岛素敏感性, 从而治疗 2型糖尿病。 如本发明所用, 术语 "丹参酮 ΠΑ" 、 "活性成分" 和 "丹参酮 ΠΑ活性物 质" 可互换使用, 是指丹参酮 ΠΑ、 其生理学可接受的盐或药学上可接受的盐、 衍生物、 水合物或它们的混合物, 其中包括但不限于: 丹参酮 ΠΑ、 其磺酸盐及 羟基丹参酮 ΠΑ。
在本发明中,对于制备丹参酮 ΠΑ的方法没有特别的限制, 比如其可以提取 自天然植物, 或者可以采用化学合成、 半化学合成的方法来制备。 丹参酮 ΠΑ的作用
丹参酮 ΠΑ在以往研究中已被应用于多种疾病的防治,例如其已被应用于对 冠心病, 脑血管和心血管疾病等疾病的防治中。 但是目前还没有关于其在防治 肥胖和 /或糖尿病和 /或缓解胰岛素抵抗方面的研究报道。 本发明人通过试验证 明了丹参酮 ΠΑ是一种天然的 PPAR y拮抗剂,由此其可作为 PPAR y拮抗剂对涉及 PPAR γ的多种疾病进行干预和治疗。 其主要作用包括但不限于以下方面:
1. 体重控制及减肥效果
为了研究丹参酮 ΠΑ的减肥效果, 本发明人首先利用高脂食物诱导肥胖小 鼠,建立肥胖模型。通过灌胃方式用丹参酮 ΠΑ对肥胖鼠进行干预。经过喂养后, 对给药组与高脂肪对照组进行外形、 体重、 生理生化等指标的比较, 验证丹参 酮 ΠΑ的减肥效果。
本发明证实了丹参酮 ΠΑ具有良好的减肥效果。 由于它是食品、动物体中天 然存在的物质, 因此安全性高, 可作为一种安全可靠的减肥活性物质。
此外, 由于丹参酮 ΠΑ活性物质具有减肥效果, 其还可用于预防、 治疗和控 制与肥胖相关的疾病, 例如但不限于: 糖尿病、 脂肪肝、 肝脏肥大、 脂质代谢 异常、 高脂血症、 动脉粥样硬化、 冠心病、 癌症、 痛风、 内分泌失调、 肺泡低 换气综合征、 胆囊炎等。 丹参酮 ΠΑ对降低血脂, 尤其是对肥胖引起的血脂过高 尤为有效。
为此,本发明提供了丹参酮 ΠΑ活性物质在制备预防、控制和治疗肥胖的药 物组合物、 食物组合物、 保健品组合物和 /或膳食添加剂中的用途。 下文将对这 些组合物进行详细描述。
此外, 在本发明中还可利用丹参酮 ΠΑ活性物质进行非治疗目的的减肥。
2. 防治糖尿病的效果
发明人通过实验惊奇地发现, 丹参酮 ΠΑ对于高脂肥胖小鼠则具有降低血 糖、 缓解胰岛素抵抗的作用, 因此可将其用于制备防治糖尿病的药物或保健品 中。
为了研究丹参酮 ΠΑ活性物质缓解胰岛素抵抗的效果,发明人对施用丹参酮 IIA后的高脂喂养小鼠的葡萄糖耐受性以及施用丹参酮 IIA的小鼠胰岛素抵抗缓 解效果进行了研究。
本发明证实了丹参酮 ΠΑ可以有效缓解胰岛素抵抗和降低血糖。由于它是天 然物质, 因此安全性高, 可作为一种安全可靠的缓解胰岛素抵抗和降低血糖的 活性物质, 从而应用于糖尿病的防治。
此外由于丹参酮 ΠΑ具有缓解胰岛素抵抗效果,其还可用于预防、治疗和控 制与胰岛素敏感性下降相关的疾病, 例如但不限于: 2型糖尿病、 高胰岛素血 症。 也可用于胰岛素抵抗导致的其它相关疾病及征状, 如: 糖尿病酮症酸中毒、 高渗性非酮症糖尿病昏迷、 乳酸性酸中毒。
为此,本发明提供了丹参酮 ΠΑ活性物质在制备预防、控制和治疗糖尿病的 疾病或征状的药物组合物、 食物组合物、 保健品组合物和 /或膳食添加剂中的用 途。 下文将对这些组合物进行详细描述。
本发明还提供了市售丹参酮 ΠΑ制剂的一种新用途, 即用于控制血糖, 缓解 胰岛素抵抗, 从而防治糖尿病。
此外,本发明的丹参酮 ΠΑ活性物质还可用于同时产生预防、控制和治疗糖 尿病和治疗肥胖的双重效果。
应理解, 本发明中所用术语"缓解"是指使得疾病或失调的征状得到改善, 甚至该疾病或失调消失或痊愈。 组合物
本发明还提供了一种组合物及其在控制体重、血糖和 /或胰岛素抵抗中的用 途,所述的组合物含有有效量的丹参酮 ΠΑ,以及食品学或药学上可接受的载体。
如本文所用, 术语 "含有" 或 "包括" 包括了 "包含" 、 "基本上由…… 构成" 、 和 "由……构成" 。
如本文所用, 术语 "药学上可接受的" 或 "食品学上可接受的" 的成分是 适用于人和 /或动物而无过度不良副反应 (如毒性、 剌激和变态反应)的, 即有合 理的效益 /风险比的物质。
如本文所用, 术语 "本发明的组合物" 包括药物组合物、 食物组合物、 保 健品组合物和 /或膳食添加剂。
如本文所用, 术语 "有效量"是指可对人和 /或动物产生功能或活性的且可 被人和 /或动物所接受的量。
如本文所用, 术语 "药学上可接受的载体" 指用于治疗剂给药的载体, 包 括各种赋形剂和稀释剂。 该术语指这样一些药剂载体: 它们本身并不是必要的 活性成分, 且施用后没有过分的毒性。 合适的载体是本领域普通技术人员所熟 知的。 在《雷明顿药物科学》(Remington' s Pharmaceutical Sciences , Mack Pub. Co. , N.J. 1991)中可找到关于药学上可接受的赋形剂的充分讨论。
本发明的组合物还可作为一种食物或膳食添加剂, 直接服用或添加到其它 食品中服用。 优选的, 所述的 "食品上学可接受的载体" 选自: 填充剂、 崩解 剂、 润滑剂、 助流剂、 泡腾剂、 矫味剂、 包覆材料、 膳食制品、 赋形剂、 或缓 /控释剂。
在组合物中药学上可接受的载体可含有液体, 如水、 盐水、 甘油和乙醇。 另外, 这些载体中还可能存在辅助性的物质, 如填充剂、 崩解剂、 润滑剂、 助 流剂、 泡腾剂、 润湿剂或乳化剂、 矫味剂、 pH缓冲物质等。 通常, 可将这些物 质配制于无毒的、 惰性的和药学上可接受的水性载体介质中, 其中 pH通常约为 5-8, 较佳地, pH约为 6-8。
本发明的组合物中可含有丹参酮 ΠΑ、 丹参酮 ΠΑ的生理学可接受的盐或药 学上可接受的盐、 水合物或它们的混合物。 本领域技术人员可根据需要确定丹 参酮 ΠΑ的服用量, 本发明丹参酮 ΠΑ组合物的每天施用量通常为 0.01-100 mg 丹 参酮 IIA/kg体重, 优选 0.05-80 mg 丹参酮 IIA/kg体重, 更优选 0.1-60 mg 丹参 酮 IIA/kg体重, 更优选 0.5-50 mg 丹参酮 IIA/kg体重, 更优选 1-25 mg 丹参酮 IIA/kg体重, 最优选 2-20 mg 丹参酮 IIA/kg体重。 。
本发明的组合物中丹参酮 ΠΑ活性物质有效成分占组合物总重量的 0.001-99.9wt%; 优选为组合物总重量的 l-95wt%, 较优选为 5-90wt%, 更优选 10-80wt%。 余量为药学上可接受的载体以及其它添加剂等物质。
如本文所用, 术语 "单位剂型" 是指为了服用方便, 将本发明的组合物制 备成单次服用所需的剂型, 包括但不限于各种固体剂 (如片剂;)、 液体剂、 胶囊 剂、 缓释剂。
在本发明的另一优选实施方式中, 所述组合物为单位剂型或多剂型, 且其 中丹参酮 ΠΑ活性物质的含量为 l-3500mg/剂。 所述组合物为单位剂型或多剂型, 且其中丹参酮 ΠΑ的含量为 l-3500mg/剂。
在本发明的一个优选例中, 所述组合物中丹参酮 ΠΑ的含量为 l-10mg/剂, 更 优选为 2-8mg/剂, 进一步优选含有 3-6mg/剂。
在本发明的另一个优选例中, 所述组合物中丹参酮 ΠΑ的含量为 10-1000mg/ 剂; 更优选 20-800mg/剂, 进一步优选 50-600mg/剂, 更优选 75-500mg/剂。
在本发明的另一个优选例中, 所述组合物中丹参酮 ΠΑ的含量为 1000-3500mg/剂; 更优选 1100-3200mg/剂, 进一步优选 1200-3000mg/剂, 更优 选 1500-2000mg/剂。
在本发明的另一个优选例中,每天施用 1-6剂本发明的组合物,优选施用 1-3 剂; 在高剂量时优选每天服用 1剂。
应理解, 所用丹参酮 ΠΑ活性物质的有效剂量可随待施用或治疗的对象的严重 程度而变化。 具体情况根据对象的个体情况 (例如对象体重、 年龄、 身体状况、 所需达到的效果;)来决定, 这在熟练医师或营养师可以判断的范围内。
本发明的组合物, 可以为固态 (如颗粒剂、 片剂、 冻干粉、 栓剂、 胶囊、 舌 下含片;)或液态 (;如口服液;)或其他合适的形状。 施用方式
本发明的组合物可以通过常规途径施用, 其中包括 (但并不限于;): 口服、 肌注、 皮下注射等。 优选口服施用。 组合物形式应与施用方式相匹配。 本发明 组合物的施用量, 按活性物质重量计, 通常为每天约 0.01- lOOmg丹参酮 IIA /kg 体重, 较佳地约 0.05-80mg 丹参酮 IIA/kg体重, 优选 0. 1-60 mg 丹参酮 IIA/kg体 重,更优选 0.5-50 mg/kg体重,更优选 1-25 mg 丹参酮 IIA/kg体重,最优选 2-20 mg 丹参酮 IIA/kg体重。
本发明的组合物可直接使用, 也可与其它治疗剂或辅剂联合使用。
在本发明的优选实施方式中, 在用于减肥或预防、 控制、 治疗与肥胖相关 的疾病中时,本发明的组合物还可与有效量 (;如 0.0005-0. 1克 /kg体重 /天;优选的, 为 0.001-0.05克 /kg体重 /天;)的选自下组的物质联合施用: 苯丙胺及其衍生物, 包 括 (但不限于;): 甲苯丙胺、 苄甲苯丙胺、 邻氯苯丙胺、 苯甲吗啉; 吲哚类药物, 包括 (但不限于 环咪吲哚、 氯苯咪吲哚; 5-羟色胺能药物, 包括 (但不限于;): 芬氟拉明、 氟西丁; e 3-AR激动剂, 包括 (但不限于): AD9677 , CP-33 U 679; 单 胺再摄取抑制剂, 包括 (但不限于;): 西部曲明; 脂酶抑制剂, 包括 (但不限于;): 奧利司他等。
在本发明的优选实施方式中, 在用于预防、 治疗与缓解糖尿病, 尤其是胰 岛素抵抗相关的疾病中, 本发明组合物还可与有效量 (;如 0.5- 100mg/kg体重 /天; 优选的, 为 l-50mg/kg体重 /天;)的选自下组的物质联合施用: 双胍类糖尿病药物 包括 (但不限于;):二甲双胍、或苯乙双胍;磺酰脲类糖尿病药物包括 (但不限于;): 格列本脲、 格列吡嗪、 格列齐持、 格列波脲、 格列美脲、 或格列喹酮; 葡萄糖 苷酶抑制剂类药物包括 (;但不限于 阿卡波糖、 伏格利波糖 (; vokibose;)、 或米格 列醇; 胰岛素增敏类药物包括 (但不限于;): 环格列酮、 曲格列酮、 罗格列酮、 或吡格列酮; 醛糖还原酶抑制剂类药物包括 (但不限于;): 阿司他丁、 依帕司他、 波拉司他、 或托瑞司他; 促胰岛素释放类药物包括 (但不限于;): 瑞格列奈、 或 那格列奈。
当两种或两种以上的药物联合给药时, 一般具有优于两种药物分别单独给 药的效果。优选地, 联合施用的药物或其它制剂不干扰本发明丹参酮 ΠΑ活性物 质有效成分的治疗活性。 非治疗目的的减肥方法
本发明还提供了一种非治疗目的的减肥方法, 所述方法包括给予对象有效 量的丹参酮 ΠΑ活性物质。
在本发明的一种优选实施方式中, 给予对象的丹参酮 ΠΑ活性物质的量为: 通常为每天约 0.001-lOOmg丹参酮 IIA /kg体重, 优选 0.01-lOOmg丹参酮 IIA /kg体 重,较佳地约 0.05-80 mg 丹参酮 IIA/kg体重,优选 0.1-60 mg 丹参酮 IIA/kg体重, 更优选 0.5-50 mg/kg体重,更优选 1-25 mg 丹参酮 IIA/kg体重,最优选 2-20 mg 丹 参酮 IIA/kg体重。 在一天中, 可以将所述的量单次给予或多次给予。
在本发明的一个优选实施方式中, 在所述减肥方法中还包括给予对象其它 减肥药物或保健品, 例如上文所述的那些减肥药物。 在本发明的另一个优选实 施方式中, 所述减肥方法中还包括其它减肥方案或措施, 例如运动、 节食等。 本发明的优点
本发明揭示了丹参酮 ΠΑ在治疗肥胖、 血糖及胰岛素敏感性中的新用途, 并由此开发了含有丹参酮 ΠΑ的新型保健品、 药品或膳食添加剂。 本发明具有 以下优点:
第一, 相对于其它防治肥胖症、 减肥和 /或糖尿病的药品或食品, 本发明 中采用的丹参酮 ΠΑ, 是食品、 动物体中天然存在的物质, 以往的研究中主要 用于食品添加剂、 合成药物等, 具有较高的安全性;
第二,丹参酮 ΠΑ在以往研究中已被应用于多种疾病的防治,例如冠心病, 脑血管和心血管疾病等疾病的防治中, 因此容易为市场所接受;
第三, 相对于已有的防治肥胖症和 /或糖尿病的保健品或药物, 丹参酮 ΠΑ 具有稳定性好、 副作用小等特点, 更加适合作为新型减肥保健品或药物进行开 发。 实施例
下面结合具体实施例, 进一步阐述本发明。 应理解, 这些实施例仅用于说 明本发明而不用于限制本发明的范围。 下列实施例中未注明具体条件的实验方 法, 通常按照常规条件, 如分子克隆实验手册(Molecular cloning : A laboratory manual , 3rd ed., Sambrook等, Cold Spring Harbor Laboratory, 2001)和植物分子生物学实验手册(Plant Molecular Biology-A Laboratory Mannual , Clark等, Springer-Verlag , 1997)中所述的条件, 或按照制造厂商 所建议的条件。
除非另外说明, 否则百分比和份数按重量计算。 除非另行定义, 文中所使 用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。 此外, 任何与 所记载内容相似或均等的方法及材料皆可应用于本发明中。 文中所述的较佳实 施方法与材料仅作示范之用。 实施例 1. 丹参酮 ΠΑ抑制脂肪细胞分化并抑制 3T3-L1细胞的基因分化
试验材料和方法
丹参酮 IIA (Tanshinone IIA, Tan)
购自浙江一新制药有限公司, 其结构如图 1A所示。
细胞培养
小鼠前脂肪细胞 3T3-L1购自 ATCC , 培养基 DMEM加 10 %胎牛血清(生长培养 基)。 脂肪细胞分化时, 将细胞传到 12孔或 24孔板, 细胞长到完全汇合后再培 养 2天, 然后用含有 10 g/ml的胰岛素、 50 μΜ的地塞米松和 0. 8 mM的 ΙΒΜΧ的生 长培养基(分化液)诱导分化。 分化 4天后分化培养基换成 DMEM加 10 %FBS的生长 培养基。 分化时细胞培养于 37 ° C , 10% C02条件中。
细胞处理 (此处所述细胞处理条件包括了整个试验过程的细胞处理条件)
用分化液诱导脂肪细胞分化的同时在分化液中加入 1 μΜ罗格列酮和 /或 5、 25、 50 μΜ的丹参酮 ΠΑ。 对于时间敏感性试验是分化的同时在分化液中加入 1 μΜ的罗格列酮, 并分别在分化的第 0天 (即分化的同时;)、 第 1天、 第 2天、 第 3天 和第 4天在分化液中加入 50 μΜ的丹参酮 ΠΑ。
油红染色
用 10%福尔马林室温固定 3T3-L1细胞 10分钟, PBS洗细胞两次然后用油红于 60° C染色 15分钟, 再用 PBS清洗两次后, 以奧林帕斯显微镜(1X71)拍照。
总 RNA提取、 cDNA制备及实时定量 PCR
组织和细胞总 RNA用 Qiagen RNA试剂盒提取。 单链 cDNA用 promega cDNA合 成试剂盒合成。
基因表达水平用 ABI 7500实时定量 PCR系统分析。 cDNA 50° C保持 2分钟, 在 95 ° C变性 10分钟后扩增 40个循环(95 ° C, 15s, 60° C, 60s)。 所有试验至少 重复 3次。 基因 mRNA水平用 β -肌动蛋白(β - actin)做内参。
免疫印记
组织和细胞直接用 2XSDS上样缓冲液裂解并煮沸 5分钟。 上样 10 总蛋白 在 10% SDS-PAGE胶上电泳后转移到 PVDF膜上。 转移后的膜用 TBS-T缓冲液配置 的 5 %脱脂奶粉溶液封闭 1小时后, 加入用 1%BSA稀释的抗体 4 ° C过夜。 上过抗 体的膜用 TBS-T缓冲液洗 3次, 每次 10分钟, 加入 HRP标记的二抗室温杂交 1小时 后再用 TBS-T洗 3次, 最后用 ECL显色液显色。
流式细胞分析
细胞生存分析试验用膜联蛋白 V-FITC/PI调亡检测试剂盒(Si gma)检测。 3T3-L1细胞收集并用膜联蛋白 V-FITC/PI染色并用 BD流式仪(BD FACSAr ia)分 析。
统计分析
试验数据用 SPSS 12. 0分析, 用单向方差分析法(AN0VA)统计分析, 当 P值 小于 0. 05时认为有统计学差异。 试验结果与分析
通过对可抑制抑制脂质形成的天然化合物的筛选,发现丹参酮 ΠΑ可以抑制 脂肪细胞的分化。
分化液诱导 3T3-L1细胞分化 5天时, 油红染色发现丹参酮 ΠΑ抑制脂肪细胞 分化呈现剂量依赖的特征(图 1B)。 丹参酮 ΠΑ在 5 μΜ时显著抑制了脂肪细胞分 化, 当浓度达到 50 μΜ时, 几乎完全抑制了脂肪细胞的分化。
正如我们所知在分化液诱导脂肪细胞分化时 PPAR γ和 C/EBPs的激活是必 须的, 无论在体内还是在体外脂质的形成都离不开 PPAR Y的激活。 为了检测丹 参酮 ΠΑ抑制脂质形成是否通过了 PPAR Y通路, 分别用实时定量 PCR和免疫印记 的方法检测了 3T3-L1细胞分化过程中 C/EBP a的 RNA和蛋白质表达水平。 结果显 示丹参酮 ΠΑ处理过的细胞 C/EBP ci水平被显著抑制, 抑制率超过 90 %, 同时, PPAR Y靶基因的表达同样被显著抑制了(图 1C, D)。
流式细胞分析结果显示丹参酮 ΠΑ的处理没有增加调亡和死亡的细胞数量, 这说明丹参酮 ΠΑ对脂肪细胞分化基因表达的抑制并不是由调亡或毒性引起的 (图 1E)。然而, 丹参酮 ΠΑ对脂肪细胞的抑制作用在分化的同时加药处理是最强 的。油红染色的结果显示在分化的第二天或更晚加入丹参酮 ΠΑ抑制效果就减弱 了,同时丹参酮 ΠΑ对 C/EBP ci的 RNA水平和蛋白水平的抑制效果也减弱了(图 1F, G和 H)。
综上所述, 研究结果提示丹参酮 ΠΑ对脂肪细胞分化的抑制作用可能通过 PPAR y通路, 并且在脂肪细胞分化的早期进行药物干预对于减少脂质形成是非 常重要的。 实施例 2.对丹参酮 ΠΑ是 PPAR γ拮抗剂的证明
试验方法
转染及报告基因分析
用于报告基因分析的质粒及试验过程如现有技术中所述(参见 Huang C等, Berberi ne inhib i ts 3T3-L1 ad ipocyte di fferent iat i on through the PPARgamma pathway. "小檗碱通过 PPAR y途径对 3T3-L1脂肪细胞分化的抑制"
Bi ochem Bi ophys Res Commun 348, 2006 : 571 -578)。 293T细胞在转染前 12小时用无抗生素的培养基铺于 48空板中, 待细胞生长 致 50 %汇合时进行转染。全长 PPAR Y表达质粒与 PPRE-J3-TK-Luc报告基因质粒 及水母荧光质粒共转到细胞中, 24小时后加入 1 μΜ罗格列酮和 /或 5、 25、 50 μΜ 的丹参酮 ΠΑ, 24小时后裂解细胞用双报告基因检测试剂盒 (; Promega)检测荧光 强度变化。 对于可以排除内源性配体的报告基因系统, Gal4— PPAR Y配体结合 域质粒与 UASG X 4-TK-Luc报告基因质粒及水母荧光质粒共转到细胞中, 加药 和分析方法同上。
闪烁迫近分析(SPA)
SPA分析是为了检测 PPAR Y与丹参酮 ΠΑ结合的亲和力的。 简言之, 生物素 标记的 PPRE与包含有鱼精 DNA, 4mg链亲合素包被的微珠的发应缓冲液混合置于 离心管中 4°C过夜培养。 混合液 1500Xg离心 10分钟。 除上清后用 10ml反应缓冲 液洗 3次。 包含有 70(^g的 ΡΡΑΙ γ蛋白, 47 人 RXRa, 10 nM 标记的罗格列 酮和不同浓度非标记罗格列酮或丹参酮 ΠΑ分别加至平底反应板中 4°C反应 4小 时后读数。 结合数据用 GraphPad Prism软件分析。 用非线性回归分析计算产生 剂量反应曲线。 值通过计算最大抑制浓度的一半 (IC )得到。
表面等离子体共振分析 (SPR)
Biacore仪器是利用 P偏振光在玻璃与金属薄膜界面处发生全内反射时渗透 到金属薄膜内的消失波,引发金属中的自由电子产生表面等离子体子的原理设 计的。 生物传感器能使非标记的生物反应信号实时传递到监视器。 结合试验是 在 25°C条件下使用 BIAcoreTM 3000仪器完成。 ΡΡΑΙ γ蛋白被固定在芯片表面。 丹参酮 ΠΑ以溶液的形式注射到芯片表面。 当丹参酮 ΠΑ与固定在芯片表面的 ΡΡΑΙ γ发生反应时, SPR反应的改变就被探测到了。 反应水平的变化与模块的 变化是等比例被记录的。
其它相关试验方法和材料如实施例 1所述。 试验结果与分析
由于丹参酮 ΠΑ可以抑制 PPAR Y通路上的基因表达, 所以推测丹参酮 ΠΑ通 过直接调控 PPAR Y活性从而抑制脂肪细胞分化。 为了检验这一观点, 向 293 Τ细 胞中共转染了一个 PPAR Y表达质粒和相应 PPAR Y信号的 PPRE荧光报告基因。 首 先验证了丹参酮 ΠΑ在罗格列酮存在和不存在的条件下都可以抑制全长的 PPAR Υ的转录活性(图 2Α)。 并且, 在可以排除内在配体干扰的 GAL4-PPAR γ LBD融 合质粒系统中, 观察到了同样的抑制效果(图 2Β)。
为了进一步证明丹参酮 ΠΑ是否是 PPAR γ的配体,设计了一套竞争性结合试 验, 在脂肪细胞分化的过程中同时加入 PPARy的强激动剂罗格列酮(1 μΜ)和丹 参酮 ΠΑ, 油红染色显示丹参酮 ΠΑ仍然可以有效抑制脂肪细胞分化(图 2C)。 在 SPA结合试验中,丹参酮 IIA可以代替放射性标记的罗格列酮与 PPARy的配 体结合结构域结合, IC5。值为 1. 62±0. 49 μΜ (¾= 1. 17±0. 35 μΜ)。 与未标记的罗 格列酮(Ki=23. 6±0· 6 ηΜ, 即 BRL49653 , 购自 Alexis Biochemicals)相比, 丹参酮
ΠΑ与 PPARy的亲和力比较适中(图 2D)。
SPR试验验证了丹参酮 ΠΑ与 PPARy的直接结合(KD, 6. 74 x 10- 5 M) (图 2E)。 报告基因试验和竞争性试验, SPA, SPR试验综合起来说明丹参酮 ΠΑ可以直接与 PPAR y 配体结合结构域结合, 由此证明了丹参酮 ΠΑ是 PPAR Y的有效拮抗剂。 实施例 3. 丹参酮 ΠΑ对高脂餐诱导的肥胖小鼠的治疗作用
试验方法
动物试验及血清生化分析
雌性 C57BL/6J小鼠购自 SLAC实验室。 所有动物饲养于 22 - 23 ° C条件下, 12小时昼夜循环。 C57BL/6J小鼠用 60 %脂肪含量的饲料喂养以诱导肥胖模型, 10 %脂肪含量的饲料喂养的小鼠用作对照。 每隔 1天的下午 1点给小鼠灌胃 35mg 丹参酮 IIA/kg体重 (200 μ 1)或者 200 μ 1 DMS0做对照。 血清甘油三酯, 总胆固 醇, 高密度脂蛋白胆固醇和低密度脂蛋白胆固醇用 Hi tachi 7020 自动分析仪 分析。
苏木精和伊红染色
白色脂肪组织用 OCT包埋, 冰冻切片法切成 10- μ ηι切片, 然后分别以苏木 精和伊红染色镜检。 试验结果与分析
过去对人和小鼠的研究显示适当的减少 PPARy活性有助于减少 2型糖尿病 的发病几率。由于丹参酮 ΠΑ是 PPAR Y的拮抗剂并且在体外可以抑制脂肪细胞分 化, 因此接下来检验丹参酮 ΠΑ在高脂餐诱导的和正常饲料喂养的 C57BL/6小鼠 的脂肪含量和体重变化中的作用。如图 3A和图 3B所示, 与对照组相比丹参酮 ΠΑ 处理可以有效预防高脂餐诱导的体重增加。并且丹参酮 ΠΑ处理组的高脂诱导的 小鼠白色脂肪组织中的脂肪细胞较之对照组的小(图 3C)。 而正常饲料喂养的小 鼠中丹参酮 ΠΑ处理组与对照组体重没有显著差异。
接着,检验丹参酮 ΠΑ是否可以减轻肥胖鼠的体重。 C57BL/6J小鼠用高脂诱 导 5个月, 体重增加到 150 %, 将肥胖小鼠分成两组, 每隔一天以丹参酮 ΠΑ (35 mg/kg)或 DMS0处理(对照)9周。 结果显示, 丹参酮 ΠΑ处理组的小鼠体重显著低 于对照组, 并且前者的脂肪细胞体积小于对照组(图 3D, E和 F)。
接着通过分析 mRNA表达水平研究丹参酮 ΠΑ在体内白色脂肪组织和肝脏中 对 PPAR Y活性及相关基因表达的影响。在白色脂肪组织中, PPAR Y的靶基因 LPL 和 UCP-2在丹参酮 IIA处理的小鼠中表达明显低于对照组, 而 SREBP- 1 , PPARa, PPARP和 S irt l没有变化, PCG- la被显著上调(图 3G)。 在肝脏中, PPAR Y的靶基 因 aP2和 LPL在丹参酮 ΠΑ处理组中明显低于对照组, 而 SREBP-1 , PPARa, PPARp 和 S i rt l仍然保持不变(图 3H)。 上述结果表明, 丹参酮 ΠΑ处理所引起的体重减 轻是通过下调 PPARy活性和其靶基因引起的。 实施例 4. 丹参酮 ΠΑ对由高脂餐诱导引起的胰岛素抵抗的作用
试验方法
腹腔注射葡萄糖耐受试验 (IPGTT)
高脂餐诱导的 C57BL/6J小鼠以丹参酮 ΠΑ或 DMS0 (对照)处理 2个月后禁食 8 小时, 腹腔注射 2 g/kg体重的葡萄糖, 在注射葡萄糖时的 0, 15, 30, 60, 120 分钟分别从尾静脉取血测量血糖检测(0分钟为未注射葡萄糖时的空腹血糖)。 免疫印记
组织和细胞直接用 2XSDS上样缓冲液裂解并煮沸 5分钟。 上样 10 总蛋白 在 10% SDS-PAGE胶上电泳后转移到 PVDF膜上。 转移后的膜用 TBS-T缓冲液配置 的 5 %脱脂奶粉溶液封闭 1小时后, 加入用 1%BSA稀释的抗体 4 ° C过夜。 上过抗 体的膜用 TBS-T缓冲液洗 3次, 每次 10分钟, 加入 HRP标记的二抗室温杂交 1小时 后再用 TBS-T洗 3次, 最后用 ECL显色液显色。
免疫酶联分析(ELISA)
为了检测丹参酮 ΠΑ对胰岛素分泌的影响, 用胰岛素 ELISA检测试剂盒 (LINCO Research)检测小鼠血清胰岛素水平。 试验结果与分析
在 2型糖尿病的早期常常会伴随高胰岛素血症, 它会引起包括高血压和充 血性心力衰竭在内的一些心血管疾病。 如图 4A和 4B所示, 高脂餐诱导的对照组 小鼠的血清胰岛素水平显著升高,而丹参酮 ΠΑ的处理有效降低了血清胰岛素水 平。 肥胖常常会引起胰岛素抵抗,后者被认为是 2型糖尿病的危险因子, 因此接 着检验丹参酮 ΠΑ处理是否可以改善由肥胖引起的胰岛素抵抗。 处理 9周的小鼠 在禁食 8小时后, 进行 IPGTT试验, 注射 2 g/kg葡萄糖后分别检测 15, 30, 60, 120分钟的血糖水平。 通过试验发现经过 5个月的高脂餐诱导, 小鼠已经发展成 为胰岛素抵抗, 而丹参酮 ΠΑ处理有效改善了胰岛素抵抗水平。
为了研究丹参酮 ΠΑ改善胰岛素抵抗提高胰岛素敏感性的机理,抽取小鼠肝 脏组织蛋白进行免疫印记试验, 检测与胰岛素抵抗相干的基因表达。 结果显示 丹参酮 ΠΑ促进了 Akt 308位苏氨酸和 473位丝氨酸的磷酸化, 同时也促进了 AMPK
172位苏氨酸及 ACC 79位丝氨酸的磷酸化(图 4D和 E)。 这些发现说明丹参酮 IIA 在体内会促进 Akt和 AMPK等在糖代谢和胰岛素敏感性中起重要作用的分子激 活。 实施例 5. 丹参酮 ΠΑ改善血脂水平
试验方法
同实施例 4。 试验结果与分析
血清生化分析结果显示在预防试验组中丹参酮 ΠΑ处理可以降低高脂餐诱 导的低密度脂蛋白及正常饮食和高脂诱导的甘油三酯水平, 而正常饮食小鼠的 高密度脂蛋白有显著提高(图 5A)。
另一方面,在治疗试验组中丹参酮 ΠΑ处理可以降低高脂餐诱导引起的甘油 三酯高密度脂蛋白和低密度脂蛋白的升高(图 5C)。 LDL-c/HDL-c的比值在所有 试验组中都有所降低(图 5B和 D)。 这些试验结果表明丹参酮 ΠΑ可以有效改善高 脂餐诱导引起的心血管损伤和高血脂风险。
在本发明提及的所有文献都在本申请中引用作为参考, 就如同每一篇文献 被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后, 本领域技术人员可以对本发明作各种改动或修改, 这些等价形式同样落于本申 请所附权利要求书所限定的范围。

Claims

权 利 要 求
1. 丹参酮 ΠΑ、其生理学上或药学上可接受的盐、其水合物或它们的混合物的 用途, 其特征在于, 用于制备预防或治疗与肥胖和 /或糖尿病相关的疾病的物质。
2. 如权利要求 1所述的用途, 其特征在于, 所述物质用于降低哺乳动物的体 重、 用于降低血脂水平和 /或用于缓解胰岛素抵抗。
3. 如权利要求 1所述的用途, 其特征在于, 所述丹参酮 ΠΑ是从天然植物提取 纯化或人工合成的。
4. 如权利要求 1所述的用途, 其特征在于, 所述物质是组合物。
5. 如权利要求 4所述的用途, 其特征在于, 所述丹参酮 ΠΑ占所述组合物总重 量的 0.001-99.9wt%。
6. 如权利要求 1所述的用途, 其特征在于, 所述物质为单位剂型或多剂型, 且其中丹参酮 ΠΑ的含量为 l-3500mg/剂。
7. 如权利要求 4所述的用途, 其特征在于, 所述组合物中还包括用于减肥、 预防和 /或治疗糖尿病的其它物质,更佳地所述的其它物质为选自下组中的一种 或多种: 甲苯丙胺、 苄甲苯丙胺、 邻氯苯丙胺、 苯甲吗啉、 环咪吲哚、 氯苯咪 吲哚、 芬氟拉明、 氟西丁、 AD9677、 CP-33 U 679、 西部曲明、 奧利司他; 二甲 双胍、 苯乙双胍、 格列本脲、 格列吡嗪、 格列齐持、 格列波脲、 格列美脲、 格 列喹酮、 阿卡波糖、 伏格利波糖、 米格列醇、 环格列酮、 曲格列酮、 罗格列酮、 吡格列酮、 阿司他丁、 依帕司他、 波拉司他、 或托瑞司他、 瑞格列奈或那格列 奈; 或它们的混合物。
8. 一种非治疗目的的减肥方法,所述方法包括给予需要减肥的对象有效量 的丹参酮 ΠΑ、 其生理学上或药学上可接受的盐、 其水合物或它们的混合物。
9. 一种组合物, 所述组合物含有:
(1) 有效量的丹参酮 ΠΑ、 其生理学上或药学上可接受的盐、 其水合物或它们 的混合物;
(2) 选自下组的一种或多种物质: 甲苯丙胺、 苄甲苯丙胺、 邻氯苯丙胺、 苯 甲吗啉、 环咪吲哚、 氯苯咪吲哚、 芬氟拉明、 氟西丁、 AD9677、 CP-33 U 679、 西部曲明、 奧利司他; 二甲双胍、 苯乙双胍、 格列本脲、 格列吡嗪、 格列齐持、 格列波脲、 格列美脲、 格列喹酮、 阿卡波糖、 伏格利波糖、 米格列醇、 环格列 酮、 曲格列酮、 罗格列酮、 吡格列酮、 阿司他丁、 依帕司他、 波拉司他、 或托 瑞司他、 瑞格列奈、 那格列奈; 或它们的混合物; 和
(3) 药学上可接受的载体或食品学上可接受的载体。
10. 丹参酮 ΠΑ、 其生理学上或药学上可接受的盐、 其水合物或它们的混合物 的用途,其特征在于,用于制备治疗通过拮抗 PPAR Y而得到缓解的疾病或失调的 物质。
PCT/CN2009/071970 2008-05-26 2009-05-26 丹参酮iia在制备治疗肥胖及胰岛素抵抗的药物中的应用 WO2009143763A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN200810038072.X 2008-05-26
CN200810038072 2008-05-26

Publications (1)

Publication Number Publication Date
WO2009143763A1 true WO2009143763A1 (zh) 2009-12-03

Family

ID=41376609

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2009/071970 WO2009143763A1 (zh) 2008-05-26 2009-05-26 丹参酮iia在制备治疗肥胖及胰岛素抵抗的药物中的应用

Country Status (2)

Country Link
CN (1) CN101590063A (zh)
WO (1) WO2009143763A1 (zh)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102304166A (zh) * 2011-07-02 2012-01-04 中国科学院昆明植物研究所 丹参酮衍生物,其药物组合物及其在制药中的应用
KR101760565B1 (ko) 2015-06-10 2017-07-31 포항공과대학교 산학협력단 Tenc1의 활성 억제제를 유효성분으로 포함하는 당뇨병성 근위축증의 예방 또는 치료용 약학적 조성물
CN113402372A (zh) * 2021-06-10 2021-09-17 广州中大南沙科技创新产业园有限公司 一种隐丹参酮衍生物及其制备方法和降脂抗肥胖的应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1482903A (zh) * 2000-10-24 2004-03-17 ֮����ʽ���� 含有那格列奈的制剂
CN1686105A (zh) * 2005-04-14 2005-10-26 刘歆 丹参酮iia磺酸钠冻干粉针及其制备方法
CN1901900A (zh) * 2003-12-30 2007-01-24 Md白奥阿尔法有限公司 用增强代谢活性的丹参衍生物治疗肥胖和代谢综合症

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1482903A (zh) * 2000-10-24 2004-03-17 ֮����ʽ���� 含有那格列奈的制剂
CN1901900A (zh) * 2003-12-30 2007-01-24 Md白奥阿尔法有限公司 用增强代谢活性的丹参衍生物治疗肥胖和代谢综合症
CN1686105A (zh) * 2005-04-14 2005-10-26 刘歆 丹参酮iia磺酸钠冻干粉针及其制备方法

Also Published As

Publication number Publication date
CN101590063A (zh) 2009-12-02

Similar Documents

Publication Publication Date Title
Diamant et al. Thiazolidinediones in type 2 diabetes mellitus: current clinical evidence
US20080081781A1 (en) Methods and compositions for the treatment of metabolic syndrome
JP5249388B2 (ja) 糖尿病の治療及び予防のためのフタリド誘導体の使用
JP2015120739A (ja) カンナビノイドの新用途
US20230285329A1 (en) Methods and Compositions for the Treatment of Steatosis-Associated Disorders
Xie et al. Dehydroabietic acid alleviates high fat diet-induced insulin resistance and hepatic steatosis through dual activation of PPAR-γ and PPAR-α
EP2405755A1 (en) Methods and compositions for the treatment of metabolic and cardiovascular disorders
US20190175583A1 (en) Pharmaceutical composition comprising amodiaquine and anti-diabetes drug as effective ingredient for prevention or treatment of diabetes
JP2019513382A (ja) 益智抽出物を有効成分として含む高尿酸血症または高尿酸血症関連の代謝障害の予防、改善または治療のための組成物
JP2020059726A (ja) 抗糖尿病及び他の有用な活性を有する植物抽出物
JP5389434B2 (ja) アディポネクチン発現低下抑制剤及びその用途
US10100011B2 (en) Pentadienoyl piperidine derivative and use thereof
WO2009143763A1 (zh) 丹参酮iia在制备治疗肥胖及胰岛素抵抗的药物中的应用
JPWO2004093910A1 (ja) PPARδアゴニストによる脳神経変性疾患治療剤
KR20200016252A (ko) 항-당뇨병성 및 다른 유용한 활성을 갖는 식물 추출물
JP6637987B2 (ja) アモジアキンを有効成分として含有する代謝性疾患の予防、改善、または治療用組成物
Jing et al. Pterostilbene ameliorates glycemic control, dyslipidemia and liver injury in type 2 diabetes rats
JP2012072136A (ja) 細胞内代謝促進用組成物、その組成物を含有する糖代謝又は脂質代謝疾患の予防及び/又は治療用医薬製剤、機能性食品及び健康食品
KR20120112973A (ko) 비만 또는 제 2형 당뇨의 예방 또는 치료용 조성물
KR101232872B1 (ko) 스핑고신-1-포스페이트(sphingosine-1-phosphate) 또는 이의 약학적으로 허용가능한 염을 유효성분으로 포함하는 비만 예방 및 치료용 약학적 조성물
WO2021037212A1 (zh) 用于对抗代谢疾病的组合物及其用途
WO2013013417A1 (zh) 具有菧类化合物结构的降血糖和减肥药物
KR20200056367A (ko) 유파틸린을 유효성분으로 포함하는 비만 예방, 치료, 또는 개선용 조성물
CN105878225B (zh) 氨基乙酰丙酸在脂肪肝治疗中的应用
CN102238952A (zh) 用2-[4-(7-乙基-5H-吡咯并[2,3-b]吡嗪-6-基)-丙-2-醇治疗肾小球性肾炎

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: 09753481

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09753481

Country of ref document: EP

Kind code of ref document: A1