WO2023221793A1 - 一种酚酸衍生物用于治疗缺血性脑卒中的应用 - Google Patents

一种酚酸衍生物用于治疗缺血性脑卒中的应用 Download PDF

Info

Publication number
WO2023221793A1
WO2023221793A1 PCT/CN2023/092591 CN2023092591W WO2023221793A1 WO 2023221793 A1 WO2023221793 A1 WO 2023221793A1 CN 2023092591 W CN2023092591 W CN 2023092591W WO 2023221793 A1 WO2023221793 A1 WO 2023221793A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
mcao
mice
reperfusion
Prior art date
Application number
PCT/CN2023/092591
Other languages
English (en)
French (fr)
Inventor
林利
王锐
罗昭依
Original Assignee
兰州大学
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 兰州大学 filed Critical 兰州大学
Publication of WO2023221793A1 publication Critical patent/WO2023221793A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/661Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C65/00Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C65/01Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups
    • C07C65/03Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups monocyclic and having all hydroxy or O-metal groups bound to the ring
    • C07C65/05Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups monocyclic and having all hydroxy or O-metal groups bound to the ring o-Hydroxy carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/12Esters of phosphoric acids with hydroxyaryl compounds

Definitions

  • the invention belongs to the fields of life sciences and medicine. Specifically, the invention relates to the application of a phenolic acid derivative in the treatment of ischemic stroke.
  • Stroke is a type of disease caused by impairment of brain blood circulation leading to loss of brain function. It is divided into ischemic stroke and hemorrhagic stroke, of which ischemic stroke accounts for up to 80%.
  • the third national retrospective sampling survey report on causes of death shows that cerebrovascular disease has become the number one cause of death among residents across the country, and stroke has the highest disability rate among single diseases.
  • the Chinese Stroke Prevention and Treatment Guidelines provide a series of guidance and suggestions.
  • thrombolytic therapy or endovascular thrombectomy is the most effective treatment for ischemic stroke.
  • the object of the present invention is to provide a compound for treating ischemic stroke or a preparation containing the compound and its mechanism of action.
  • the present invention provides the action mechanism and application of a class of phenolic acid derivatives, or pharmaceutically acceptable salts thereof, or solvates thereof, or prodrugs thereof, in the treatment of ischemic stroke. That is, phenolic acid derivatives can inhibit the proliferation of astrocytes and microglia, reduce the expression levels of pro-inflammatory factors (TNF- ⁇ , IL-1 ⁇ , IL-6, iNOS, COX2) and HIF-1 ⁇ . Inhibiting the occurrence and development of inflammation can achieve the effect of treating ischemic stroke and provide a new method for the treatment of ischemic stroke.
  • pro-inflammatory factors TNF- ⁇ , IL-1 ⁇ , IL-6, iNOS, COX2
  • a compound represented by Formula I below or a pharmaceutically acceptable salt thereof, an optical isomer thereof, a hydrate, a solvate or a prodrug;
  • X is selected from the following group: O or S;
  • R 1 and R 2 are each independently selected from the group consisting of: OH, SH, NH 2 , X 2 -PO(OH) 2 , or X 2 -PS(OH) 2 ;
  • X 2 is selected from the following group; O or S;
  • It is characterized in that it is used to prepare a pharmaceutical composition for treating and/or alleviating ischemic stroke.
  • the compound of formula I has a structure selected from the following group:
  • X is defined as mentioned above.
  • the compound of formula I has a structure selected from the following group:
  • the compound of formula I has a structure selected from the following group:
  • X is defined as mentioned above.
  • the compound of formula I is selected from the following group:
  • the pharmaceutically acceptable salt is selected from the following group: alkali metal salts, alkaline earth metal salts, and ammonium salts.
  • the pharmaceutically acceptable salt of the compound of formula I is the pentaammonium salt of the compound of formula I.
  • the pharmaceutical composition is also used to inhibit the proliferation of astrocytes and microglia.
  • the pharmaceutical composition is used to improve and/or alleviate the inflammatory response caused by ischemic stroke.
  • the pharmaceutical composition is also used to reduce the expression level of pro-inflammatory factors.
  • the pro-inflammatory factor is selected from the following group: TNF- ⁇ , IL-1 ⁇ , IL-6, iNOS or COX2.
  • the pharmaceutical composition is also used to reduce the expression level of oxygen homeostasis regulatory factor HIF-1 ⁇ .
  • the pharmaceutical composition improves stroke symptoms by down-regulating the expression level of the oxygen homeostasis regulatory factor HIF-1 ⁇ .
  • a method for treating and/or alleviating ischemic stroke is provided, which is characterized by comprising the steps of: administering a safe and effective amount of Compound 2 to a subject in need, or a pharmaceutically acceptable amount thereof salts, optical isomers, hydrates, solvates or prodrugs thereof.
  • Figure 1 shows the construction of MCAO model mice.
  • A TTC staining results of mouse brain tissue after one day of sham operation and cerebral ischemia-reperfusion.
  • B Statistical results of TTC staining results of brain tissue.
  • C Laser speckle detection of cerebral blood flow.
  • D Cerebral blood flow index statistical results.
  • E Statistical results of Zea-Longa neurobehavioral score of mice after one day of sham operation and ischemia-reperfusion.
  • Figure 2 shows the effect of compound 2 on the neurological function scores of MCAO model mice.
  • Figure 3 shows the effect of compound 2 on ischemic infarct volume in MCAO mice.
  • A TTC staining of mouse brains in model group and compound 2 group.
  • B Statistics of ischemic infarction area of mice in model group and compound 2 group.
  • Figure 4 shows the effects of compound 2 on the balance, neurological damage, body weight and survival rate of model mice.
  • A Statistical results of the balanced rotating rod experiment.
  • B Neurologic impairment on a fourteen-point scale.
  • C Statistical results of mouse body weight change rate.
  • D Mouse survival curve after ischemia-reperfusion.
  • Figure 5 shows the results of GFAP immunofluorescence after ischemia-reperfusion.
  • B Statistical results of GFAP-positive cell density in the brain tissue of the model group (MCAO), compound 2 group, and compound 1 group.
  • Figure 6 shows the expression of GFAP protein in each group after ischemia-reperfusion.
  • A Representative pictures of Western blot (WB) of GFAP protein expression in the brain tissue of each group (SHAM), model group (MCAO), compound 2 group, and compound 1 group;
  • B Gray WB of GFAP protein in each group Degree statistical results.
  • Figure 7 shows the results of iba1 immunofluorescence after ischemia-reperfusion.
  • B Statistical results of iba1-positive cell density in the brain tissue of the model group (MCAO), compound 2 group, and compound 1 group after 72 hours of ischemia-reperfusion.
  • Figure 8 shows the expression of iba1 protein in each group after ischemia-reperfusion.
  • A Representative WB pictures of iba1 protein expression in the brain tissue of the sham operation group (SHAM), model group (MCAO), compound 2 group, and compound 1 group.
  • B Statistical results of gray value of iba1 protein WB in each group.
  • Figure 9 shows the results of TNF- ⁇ immunofluorescence after ischemia-reperfusion.
  • B Statistical results of TNF- ⁇ positive cell density in the brain tissue of the model group (MCAO), compound 2 group, and compound 1 group.
  • Figure 10 shows the results of IL-1 ⁇ immunofluorescence after ischemia-reperfusion.
  • B Statistical results of IL-1 ⁇ positive cell density in the brain tissue of the model group (MCAO), compound 2 group, and compound 1 group.
  • Figure 11 shows the results of IL-6 immunofluorescence after ischemia-reperfusion.
  • B Statistical results of IL-6 positive cell density in the brain tissue of the model group (MCAO), compound 2 group, and compound 1 group.
  • Figure 12 shows the results of iNOS immunofluorescence after ischemia-reperfusion.
  • B Statistical results of iNOS positive cell density in the brain tissue of the model group (MCAO), compound 2 group, and compound 1 group.
  • Figure 13 shows the results of COX2 immunofluorescence after ischemia-reperfusion.
  • B Statistical results of COX2-positive cell density in the brain tissue of the model group (MCAO), compound 2 group, and compound 1 group.
  • Figure 14 shows the expression of pro-inflammatory factor mRNA in each group after ischemia-reperfusion.
  • Figure 15 shows the expression of TNF- ⁇ , IL-1 ⁇ , and IL-6 in brain tissue after ischemia-reperfusion measured by enzyme-linked immunosorbent assay.
  • the expression levels of TNF- ⁇ (A), IL-1 ⁇ (B), and IL-6 (C) in the brain tissue homogenates of the sham operation group (SHAM), model group (MCAO), compound 2 group, and compound 1 group were related to statistical results.
  • Figure 16 shows the expression of TNF- ⁇ protein in each group after ischemia-reperfusion.
  • A WB representative pictures of TNF- ⁇ protein expression in brain tissue of each group (SHAM), model group (MCAO), compound 2 group, and compound 1 group;
  • B Gray WB of TNF- ⁇ protein in each group Degree statistical results.
  • Figure 17 shows the results of HIF-1 ⁇ immunofluorescence after ischemia-reperfusion.
  • B Statistical results of HIF-1 ⁇ positive cell density in the brain tissue of the model group (MCAO), compound 2 group, and compound 1 group.
  • Figure 18 shows the mRNA expression of HIF-1 ⁇ after ischemia-reperfusion.
  • Figure 19 shows the expression of HIF-1 ⁇ protein in each group after ischemia-reperfusion.
  • A Representative WB pictures of HIF-1 ⁇ protein expression in brain tissues of the sham operation group (SHAM), model group (MCAO), compound 2 group, and compound 1 group.
  • B Statistical results of gray value of HIF-1 ⁇ protein WB in each group.
  • a compound with the structure shown in Formula I, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a prodrug thereof is an effective treatment for ischemia.
  • Active ingredient in stroke Experiments have shown that compounds represented by formula I can inhibit the proliferation of astrocytes and microglia, and reduce pro-inflammatory factors (TNF- ⁇ , IL-1 ⁇ , IL-6, iNOS, COX2) and HIF-1 ⁇
  • TNF- ⁇ , IL-1 ⁇ , IL-6, iNOS, COX2 pro-inflammatory factors
  • HIF-1 ⁇ The expression level can achieve the effect of treating ischemic stroke by inhibiting the occurrence and development of inflammation.
  • an active ingredient that can treat ischemic stroke is provided.
  • the active ingredient is a compound represented by formula (I), or a pharmaceutically acceptable salt thereof, an optical isomer, a hydrate, a solvate or a prodrug thereof;
  • X is selected from the following group: O or S;
  • R 1 and R 2 are each independently selected from the group consisting of: OH, SH, NH 2 , X 2 -PO(OH) 2 , or X 2 -PS(OH) 2 ;
  • X 2 is selected from the following group; O or S;
  • the preferred compound is compound 1 or compound 2:
  • the active ingredients of the present invention can inhibit the proliferation of astrocytes and microglia, and reduce the expression of pro-inflammatory factors (TNF- ⁇ , IL-1 ⁇ , IL-6, iNOS, COX2) and HIF-1 ⁇ level, by inhibiting the occurrence and development of inflammation to achieve the effect of treating ischemic stroke.
  • active ingredient As used herein, “active ingredient”, “active compound of the present invention” and “active ingredient of the present invention” are used interchangeably and all refer to the compound of Formula I of the present invention and its structural analogs.
  • the active ingredient of the present invention includes the compound of formula I of the present invention, or a pharmaceutically acceptable salt, enantiomer, diastereomer or racemate thereof, or a prodrug thereof. It should be understood that the active ingredient of the present invention also includes various crystal forms, amorphous compounds, deuterated compounds and other forms of the compound of formula I of the present invention.
  • the "pharmaceutically acceptable salt” is a sodium salt, potassium salt, calcium salt, aluminum salt or ammonium salt formed by a compound of formula (I) and an inorganic base; or a methylamine salt formed by a compound of formula (I) and an organic base.
  • a preferred type of pharmaceutically acceptable salt is an ammonium salt, more preferably a pentaammonium salt.
  • the present invention also provides a compound of formula I, or a mixture of one or more of the pharmaceutically acceptable salts, enantiomers, diastereomers or racemates and prodrugs thereof as active ingredients.
  • the pharmaceutical composition provided by the present invention preferably contains active ingredients in a weight ratio of 0.001-99wt%.
  • the preferred ratio is that the compound of formula I as the active ingredient accounts for 0.1wt%-90wt% of the total weight, and the remainder is a pharmaceutically acceptable carrier. , diluent or solution or salt solution.
  • the carrier includes conventional diluents, excipients, fillers, binders, wetting agents, disintegrants, absorption accelerators, surfactants, adsorption carriers, lubricants, etc. in the pharmaceutical field.
  • the compounds and pharmaceutical compositions provided by the present invention can be in various forms, such as tablets, capsules, powders, syrups, solutions, suspensions and aerosols, etc., and can be present in suitable solid or liquid carriers or diluents. Neutralize suitable for use in sterile equipment for injection or infusion.
  • the unit dosage of the preparation formula usually contains 0.05-400 mg of the compound of formula (I).
  • the unit dosage of the preparation formula contains 1 mg-500 mg of the compound of formula (I).
  • the compounds and pharmaceutical compositions of the present invention can be used clinically on mammals, including humans and animals, through The route of administration is through the mouth, nose, skin, lungs or gastrointestinal tract, and injection preparations (such as infusions) are most preferred.
  • the most preferred daily dose is 0.01-400 mg/kg body weight, taken at one time, or 0.01-200 mg/kg body weight taken in divided doses. Regardless of the method of administration, the optimal dose for the individual will depend on the specific treatment. Usually, you start with a small dose and gradually increase the dose until you find the most suitable dose.
  • the drugs or inhibitors of the present invention can be administered in various ways, for example, through injection, spray, nasal drop, eye drop, penetration, absorption, physical or chemical mediated method, such as muscle, intradermal, subcutaneous, intravenous introduction into the body. , mucosal tissue; or mixed or wrapped with other substances and introduced into the body.
  • the compound of formula I of the present invention can significantly improve cerebral infarction caused by ischemia-reperfusion, significantly improve the movement coordination and balance of ischemia-reperfusion mice, significantly improve their neurological damage, and can also significantly inhibit the The decrease in body weight reduces the mortality of mice.
  • the compound of the present invention has low toxic and side effects and good medicinal properties.
  • mice Male C57BL/6 mice were used for research and brain tissue extraction. All animals used in this experiment were obtained from the Lanzhou Veterinary Research Institute of the Chinese Academy of Agricultural Sciences. Animals were raised under standard conditions: room temperature (20 ⁇ 2°C) and a 12-h day-night cycle. Experimental animals were supplied with water and standard feed, and were acclimated for 3 days before conducting experiments. All animal experiments will be conducted from 8 a.m. to 6 p.m. The animal experiments involved in this study strictly followed the Lanzhou University Experimental Animal Ethics Code (License Number: JCYXY Gan 2021-0126).
  • Compound 1 and compound 2 were prepared according to the method described in Liu Huiyun. Design, synthesis and activity evaluation of new blood oxygen regulators [D]. Lanzhou University, 2019.
  • Example 1 Establishment and verification of middle cerebral artery occlusion (MCAO) model
  • This experiment uses the method of Longa et al. to construct a mouse MCAO model.
  • This model can well simulate the occurrence, development and acute phase treatment of ischemic stroke in clinical practice, and can provide guidance for the prevention and treatment of ischemic stroke.
  • Mechanism research provides stable application value.
  • mice were tested The brain infarct volume on the first day after surgery, the experimental results are shown in Figure 1A and Figure 1B, stained with 2,3,5-triphenyte-trazoliumchloride (TTC) The results showed that brain infarction occurred one day after surgery, and the proportion of infarct volume was 45.14 ⁇ 4.48% (P ⁇ 0.001).
  • mice weighing 23 ⁇ 1g were randomly divided into sham operation group (SHAM), model group (MCAO) and drug administration group (50mg/kg, 100mg/kg, 200mg/kg) with 6 mice each.
  • SHAM sham operation group
  • MCAO model group
  • drug administration group 50mg/kg, 100mg/kg, 200mg/kg
  • mice in the SHAM group were separated from blood vessels, but no thread plugs were released, and no treatment was given before and after surgery; the mice in the model group were constructed to construct an ischemic MCAO model, and no treatment was given before and after surgery; the administration group was treated with Compound 2 on the basis of the model group.
  • Mice with a neurofunctional score of 0 after reperfusion and mice that died before 72 h after surgery were excluded.
  • the drugs were administered at 0 h, 1 day and 2 days after reperfusion.
  • the administration method was tail vein administration.
  • the SHAM group and the MCAO group were given equal volumes of normal saline
  • the neurological function score reflects the neurological injury status of MCAO model mice.
  • the neurological function score was performed 24 hours after modeling (refer to the Zea-Longa score screening method). Mice with a score of 1-3 are considered successful in modeling, and those with a score of 0 or 4 will be eliminated.
  • the scoring criteria are as follows:
  • the sham operation group (SHAM), model group (MCAO), compound 2 (50mg/kg) group, compound 2 (100mg/kg) group and compound 2 (200mg/kg) group were evaluated by Zea-Longa scoring method.
  • the main scoring time points are preoperative, 1 day, 2 days, and 3 days.
  • the experimental results are shown in Figure 2.
  • the score of the sham operation group was 0, indicating that the mice did not suffer neurological damage.
  • the mice in the MCAO group and compound 2 group showed circles to one side at 24 hours of reperfusion. Obvious phenomenon indicates that the model is successfully constructed.
  • Example 3 Effect of compound 2 on cerebral infarct volume in MCAO model mice
  • mice of the sham operation group (SHAM), the model group (MCAO), the compound 2 (50 mg/kg) group, the compound 2 (100 mg/kg) group and the compound 2 (200 mg/kg) in Example 2 were detected by TTC staining.
  • the present invention takes Compound 1 and Compound 2 as examples to study the mechanism of the compounds of the present invention in treating ischemic stroke.
  • mice weighing 23 ⁇ 1g were randomly divided into sham operation group (SHAM), model group (MCAO), compound group 2 (100mg/kg) and compound group 1 (50mg/kg ) 10 each.
  • SHAM sham operation group
  • MCAO model group
  • compound group 2 100mg/kg
  • compound group 1 50mg/kg
  • the mice in the SHAM group were separated from blood vessels, but no thread plugs were released, and no treatment was given before and after surgery; the mice in the model group were constructed as MCAO models, and no treatment was given before and after surgery; the administration group was given Compound 2 or Compound 2 on the basis of the model group. 1 treatment.
  • Mice with a neurofunctional score of 0 after reperfusion and mice that died before 72 h after surgery were excluded.
  • the administration time for the animal behavior test was seven times at 0 h, 1 day, 2 days, 3 days, 4 days, 5 days, and 6 days after reperfusion.
  • the administration time for other experiments was at 0 h, 1 day, and 2 days after reperfusion. Dosing three times a day.
  • the administration method was tail vein administration, and the sham operation group and the model group were given equal amounts of normal saline.
  • Example 4 Effects of the compounds of the present invention on behavioral functions of MCAO model mice
  • mice The motor coordination of mice was tested using a balancing rotarod.
  • the experiment was divided into four groups: sham operation group (SHAM), model group (MCAO), compound 2 group and compound 1 group.
  • SHAM sham operation group
  • MCAO model group
  • compound 2 group The effect of compound 2 on the recovery of behavioral functions of mice after ischemia-reperfusion was comprehensively analyzed.
  • the rotarod test can test the motor coordination ability of mice.
  • the experimental program is set to gradually accelerate the speed of the rotary rod from 10 rpm to 40 rpm within 300 seconds. Record the time the mouse stays on the rotarod. Mice were trained 3 days in advance. Select mice that can stay on the rotating rod for about 300 seconds for the next experiment. The time the mouse stayed on the rotarod before constructing the model was selected as the preoperative test value.
  • mice in the SHAM group, MCAO group, compound 2 group, and compound 1 group were tested on the rotarod, and the time the mice stayed on the rotarod was recorded.
  • mice neurological deficit score (mNSS) table (14-point scale)
  • mice in the modified mouse neurological deficit score (mNSS) method shown in the table above was used to perform neurofunctional scores (14-point scale) on mice in each group.
  • Neurobehavioral scores were performed on the mice in the SHAM group, MCAO group, compound 2 group, and compound 1 group on days 1, 3, 5, and 7 after surgery.
  • Mice with scores of 10 to 14 indicated severe neurological damage.
  • Mice with a score of 5 to 9 indicated moderate neurological damage.
  • a score of 1 to 4 indicates mild damage to the nervous system.
  • the maximum score is 14 points.
  • Analysis of the neurological damage score results is shown in Figure 4B.
  • the neurological function of mice in the Compound 2 group was significantly improved from 1 day to 7 days of reperfusion.
  • the nerve function of compound 2 group was significantly improved.
  • Example 5 Effect of the compound of the present invention on astrocytes in MCAO model mice
  • Example 6 Effect of the compounds of the present invention on microglial activation and proliferation in MCAO model mice
  • Microglia are resident phagocytes in the nervous system and are the primary defense system against stress and damage to central nervous cells. After cerebral ischemia-reperfusion, microglia are activated after external injury stimulation and release pro-inflammatory mediators and anti-inflammatory mediators.
  • iba1 ionized calcium binding adapter molecule1
  • SHAM sham operation group
  • MCAO model group
  • compound 2 group compound 1 group. Fluorescence expression of iba1 in tissues.
  • iba1 a specific protein of microglia in brain tissue
  • SHAM sham operation group
  • MCAO model group
  • compound 2 group compound 1 group
  • WB representative picture of protein expression is shown in Figure 8A
  • Figure 8B The results showed that compared with the sham operation group, the expression of iba1 protein in the brain tissue of mice in the MCAO model group was significantly increased by 110 ⁇ 50% (P ⁇ 0.001). Compared with the MCAO group, three days after compound 2 was administered, the expression of iba1 protein was significantly reduced by 50 ⁇ 12% (P ⁇ 0.05).
  • Example 7 Effects of the compounds of the present invention on the expression levels of inflammatory factors in MCAO model mice
  • ischemic stroke The exact cause of ischemic stroke is still unclear, but research shows that inflammatory response plays an important role in the pathogenesis of ischemic stroke.
  • the expression changes of inflammatory factors can be used as effective evaluation indicators for the therapeutic effect of ischemic brain injury.
  • the infiltration of blood-borne leukocytes into the brain parenchyma and the activation of endogenous microglia are the reasons for the strong inflammatory response after cerebral ischemia.
  • microglia and infiltrating immune cells are significantly increased in brain tissue.
  • Microglia promote neuroinflammation by releasing pro-inflammatory factors (TNF- ⁇ , IL-1 ⁇ , IL-6, etc.) and cytotoxic molecules (IFN- ⁇ , prostaglandins, ROS, NO, etc.).
  • Microglia may cause neuronal damage through the following mechanisms: 1) secretion of pro-inflammatory factors; 2) activation of nitrogen oxides (NOX), leading to microglial proliferation and neuroinflammation; 3) expression of iNOS; 4) damage to neurons of endocytosis.
  • Cyclooxygenase (COX) plays a key role in prostaglandin biosynthesis.
  • COX2 as a dioxygenase and peroxidase, mediates the formation of prostaglandins from arachidonic acid and plays a key role in the occurrence of inflammation.
  • COX2 is induced to express when stimulated by specific events, such as producing prostaglandins in response to physiological stress such as infection and inflammation.
  • this study used immunofluorescence experiments to observe the expression of TNF- ⁇ , IL-1 ⁇ , IL-6, iNOS and COX2 proteins in brain tissue, and RT-qPCR experiments.
  • the mRNA gene expression of pro-inflammatory factors TNF- ⁇ , IL-1 ⁇ , IL-6, iNOS and COX2 in the sham operation group, model group, compound 2 group and compound 1 group was detected.
  • the concentrations of TNF- ⁇ , IL-1 ⁇ and IL-6 in brain tissue homogenate were detected by enzyme-linked immunosorbent assay (ELISA), and the brain tissue was detected by Western blot.
  • ELISA enzyme-linked immunosorbent assay
  • the number of pro-inflammatory factor-positive cells also decreased significantly after three days of compound 1 administration (P ⁇ 0.001). Compared with compound 1 group, the density of pro-inflammatory factor-positive cells in compound 2 group was significantly reduced (P ⁇ 0.001). As shown in Figure 9, after MCAO model mice were treated with Compound 2, the number of TNF- ⁇ -positive cells decreased significantly (140 ⁇ 35 vs 361 ⁇ 48/mm 2 , P ⁇ 0.001), and the number of TNF- ⁇ -positive cells in Compound 1 Although the number of cells decreased (244 ⁇ 36 vs 361 ⁇ 48/mm 2 , P ⁇ 0.001), the inhibitory effect was not as significant as that of compound 2 group.
  • RT-qPCR experimental results and quantitative analysis results are shown in Figure 14.
  • the mRNA expression of TNF- ⁇ , IL-1 ⁇ , IL-6, iNOS and COX2 in brain tissue was measured by RT-qPCR. Detection and analysis. The results showed that compared with the sham operation group, the MCAO group had higher levels of TNF- ⁇ (P ⁇ 0.001), IL-1 ⁇ (P ⁇ 0.001), IL-6 (P ⁇ 0.01), iNOS (P ⁇ 0.001) and COX2 (P ⁇ 0.001) expression levels increased to varying degrees.
  • TNF- ⁇ (P ⁇ 0.05), IL-1 ⁇ (P ⁇ 0.01), IL-6 (P ⁇ 0.01), iNOS (P ⁇ 0.05) and COX2 (P ⁇ 0.001) expression level was significantly reduced.
  • compound 2 had the most obvious inhibitory effect on the expression of IL-6, which decreased by 51 ⁇ 8%.
  • the expressions of TNF- ⁇ (P ⁇ 0.05), iNOS (P ⁇ 0.05) and COX2 (P ⁇ 0.001) were significantly reduced to varying degrees.
  • n 3
  • the data are mean ⁇ SEM, one-way ANOVA analysis method is used for processing analysis, and Tukey's HSD test is performed.
  • the ELISA experimental results and quantitative analysis results are shown in Figure 15.
  • the expression levels of inflammatory factors TNF- ⁇ , IL-1 ⁇ , and IL-6 in the mouse brain tissue homogenate were detected.
  • the results showed that compared with the sham operation group, the expression of TNF- ⁇ (P ⁇ 0.01), IL-1 ⁇ (P ⁇ 0.001), and IL-6 (P ⁇ 0.01) in the MCAO group increased to varying degrees.
  • the expressions of TNF- ⁇ , IL-1 ⁇ and IL-6 were significantly reduced, among which the expression of TNF- ⁇ was reduced by 40 ⁇ 10% (P ⁇ 0.01).
  • IL-1 ⁇ was reduced by 32 ⁇ 9% (P ⁇ 0.001), and the expression of IL-6 was reduced by 40 ⁇ 10% (P ⁇ 0.05).
  • the expression level of TNF- ⁇ protein in brain tissue was detected by Western blot.
  • the experimental and quantitative analysis results are shown in Figure 16.
  • the protein expression of TNF- ⁇ in the brain tissue of mice in the MCAO group was significantly increased by 120 ⁇ 50% (P ⁇ 0.001).
  • the expression of TNF- ⁇ protein was significantly reduced by 40 ⁇ 12% (P ⁇ 0.001).
  • the expression of TNF- ⁇ protein decreased by 20 ⁇ 10% (P ⁇ 0.001).
  • the protein expression of TNF- ⁇ in compound 2 group decreased by 31 ⁇ 20% (P ⁇ 0.05).
  • Example 8 Effect of compounds of the present invention on hypoxia-inducible factors in MCAO model mice
  • HIF-1 ⁇ is a sensitive oxygen homeostasis regulator whose expression is rapidly induced after hypoxia-ischemia. It plays a broad role in the pathophysiology of stroke, including neuronal survival, neuroinflammation, angiogenesis, glucose metabolism, and blood-brain barrier regulation. It can not only produce specific feelings when the body is hypoxic, but also facilitate the maintenance of the body's oxygen homeostasis.
  • This study used immunofluorescence experiments to observe the expression level of HIF-1 ⁇ in brain tissue, Western blot to detect the expression level of HIF-1 ⁇ in brain tissue, and RT-qPCR experiments to detect the sham operation group, model group, compound 2 group and compound Gene expression of related factors in group 1.
  • HIF-1 ⁇ protein expression in the compound 2 group was significantly reduced.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明提供了一种酚酸衍生物用于治疗缺血性脑卒中的应用,具体地,本发明提供了一种如下式(I)所示的化合物,或其药学上可接受的盐、其光学异构体,水合物、溶剂化物或前药的用途,其用于制备用于治疗和/或缓解缺血性脑卒中的药物组合物。

Description

一种酚酸衍生物用于治疗缺血性脑卒中的应用 技术领域
本发明属于生命科学和医药领域,具体地,本发明涉及一种酚酸衍生物在治疗缺血性脑卒中中的应用。
背景技术
脑卒中是由脑部血液循环发生障碍导致大脑功能缺失的一类疾病,分为缺血性脑卒中和出血性脑卒中,其中缺血性脑卒中所占比例高达80%。
全球范围内,有六分之一左右的人会发生脑卒中,每年脑卒中新发病例超过1500万。每年有500万患者由于脑卒中而失去生命,还有500万患者由于脑卒中造成终身残疾而失去生活自理能力,只有少部分人能够得到较好的治疗。
全国第三次死因回顾抽样调查报告显示,脑血管疾病已经成为全国居民死亡病因的首位,脑卒中则是单病种致残率最高的疾病。针对缺血性脑卒中的预防与治疗,中国脑卒中防治指导规范给出一系列指导建议,目前溶栓治疗或血管内取栓是缺血性脑卒中最有效的治疗方式。即便如此,缺血性脑卒中的治疗仍然存在诸多问题急需解决。第一,很多患者在发病后错过治疗时间窗没有进行及时的治疗;第二,虽然部分患者能够在短暂的治疗时间窗口内得到就医,但是大部分患者在进行治疗后仍会留下不同程度残疾,还有少部分患者由于缺血再灌注损伤而导致病情加重;第三,目前仍然没有针对缺血性脑卒中后遗症的有效治疗方法。因此,持续深入地研究缺血性脑卒中疾病的相关分子机制,寻找和研发新型抗缺血性脑卒中药物具有潜在的应用价值和重要的研究意义。
综上所述,本领域还需要开发适用于治疗缺血性脑卒中的药物。
发明内容
本发明的目的是提供一种治疗缺血性脑卒中的化合物或含有所述化合物的制剂及其作用机制。
具体地,本发明提供了一类酚酸衍生物、或其药学上可接受的盐、或其溶剂化物、或其前药在治疗缺血性脑卒中的作用机制和应用。即酚酸衍生物能够抑制星形胶质细胞与小胶质细胞的增生,降低促炎因子(TNF-α、IL-1β、IL-6、iNOS、COX2)及HIF-1α的表达水平,通过抑制炎症的发生发展从而达到治疗缺血性脑卒中的效果,为缺血性脑卒中的治疗提供新的方法。
在本发明的第一方面,提供了一种如下式I所示的化合物,或其药学上可接受的盐、其光学异构体,水合物、溶剂化物或前药的用途;
其中,X选自下组:O或S;
R1和R2各自独立地选自下组:OH、SH、NH2、X2-PO(OH)2、或X2-PS(OH)2
X2选自下组;O或S;
表示
其特征在于,用于制备用于治疗和/或缓解缺血性脑卒中的药物组合物。
在另一优选例中,所述的式I化合物具有选自下组的结构:
其中,X的定义如上文中所述。
在另一优选例中,所述的式I化合物具有选自下组的结构:
在另一优选例中,所述的式I化合物具有选自下组的结构:
其中,X的定义如上文中所述。
在另一优选例中,所述的式I化合物选自下组:
在另一优选例中,所述的药学上可接受的盐选自下组:碱金属盐、碱土金属盐、铵盐。
在另一优选例中,所述式I化合物的药学上可接受的盐为式I化合物的五铵盐。
在另一优选例中,所述的药物组合物还用于抑制星形胶质细胞与小胶质细胞的增生。
在另一优选例中,所述的药物组合物用于改善和/或缓解缺血性脑卒中引起的炎症反应。
在另一优选例中,所述的药物组合物还用于降低促炎因子的表达水平。
在另一优选例中,所述的促炎因子选自下组:TNF-α、IL-1β、IL-6、iNOS或COX2。
在另一优选例中,所述的药物组合物还用于降低氧稳态调节因子HIF-1α的表达水平。
在另一优选例中,所述的药物组合物通过下调氧稳态调节因子HIF-1α的表达水平从而改善脑卒中症状。
在本发明的另一方面,提供了一种治疗和/或缓解缺血性脑卒中的方法,其特征在于,包括步骤:给需要的对象施用安全有效量的化合物2,或其药学上可接受的盐、其光学异构体,水合物、溶剂化物或前药。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
附图说明
图1是MCAO模型小鼠的构建。(A)假手术与脑缺血再灌注一天后小鼠脑组织TTC染色结果。(B)脑组织TTC染色结果统计结果。(C)激光散斑检测脑血流。(D)脑血流指数统计结果。(E)假手术与缺血再灌注一天后小鼠Zea-Longa神经行为学评分统计结果。
图2是化合物2对MCAO模型小鼠神经功能评分的影响。
图3是化合物2对MCAO小鼠缺血梗死体积影响。(A)模型组和化合物2组小鼠脑TTC染色。(B)模型组和化合物2组小鼠缺血梗死面积的统计。
图4是化合物2对模型小鼠的平衡、神经功能损伤、体重和生存率的影响。(A)平衡转棒实验统计结果。(B)十四分制的神经功能损伤。(C)小鼠体重变化率统计结果。(D)缺血再灌注后小鼠生存曲线。
图5是缺血再灌注后GFAP免疫荧光结果。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中GFAP的荧光染色代表图片。(Scale bar=100μm)。(B)模型组(MCAO)、化合物2组、化合物1组各组脑组织中GFAP的阳性细胞密度统计结果。
图6是缺血再灌注后各组GFAP蛋白的表达情况。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中GFAP蛋白表达的Western blot(WB)代表图片;(B)各组GFAP蛋白WB的灰度值统计结果。
图7是缺血再灌注后iba1免疫荧光结果。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中iba1的荧光染色代表图片。(Scale bar=100μm)。(B)缺血再灌注72h后的模型组(MCAO)、化合物2组、化合物1组各组脑组织中iba1阳性细胞密度统计结果。
图8是缺血再灌注后各组iba1蛋白表达情况。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中iba1蛋白表达的WB代表图片。(B)各组iba1蛋白WB的灰度值统计结果。
图9是缺血再灌注后TNF-α免疫荧光结果。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中TNF-α的荧光染色代表图片。(Scale bar=100μm)。(B)模型组(MCAO)、化合物2组、化合物1组各组脑组织中TNF-α的阳性细胞密度统计结果。
图10是缺血再灌注后IL-1β免疫荧光结果。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中IL-1β的荧光染色代表图片。(Scale bar=100μm)。(B)模型组(MCAO)、化合物2组、化合物1组各组脑组织中IL-1β的阳性细胞密度统计结果。
图11是缺血再灌注后IL-6免疫荧光结果。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中IL-6的荧光染色代表图片。(Scale bar=100μm)。(B)模型组(MCAO)、化合物2组、化合物1组各组脑组织中IL-6的阳性细胞密度统计结果。
图12是缺血再灌注后iNOS免疫荧光结果。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中iNOS的荧光染色代表图片。(Scale bar=100μm)。(B)模型组(MCAO)、化合物2组、化合物1组各组脑组织中iNOS的阳性细胞密度统计结果。
图13是缺血再灌注后COX2免疫荧光结果。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中COX2的荧光染色代表图片。(Scale bar=100μm)。(B)模型组(MCAO)、化合物2组、化合物1组各组脑组织中COX2的阳性细胞密度统计结果。
图14是缺血再灌注后各组促炎因子mRNA表达情况。假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中TNF-α(A)、IL-1β(B)、IL-6(C)、iNOS(D)和COX2(E)的mRNA表达量与统计结果。
图15是酶联免疫法测定缺血再灌注后脑组织中TNF-α、IL-1β、IL-6表达情况。假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织匀浆液中TNF-α(A)、IL-1β(B)、IL-6(C)表达量与统计结果。
图16是缺血再灌注后各组TNF-α蛋白的表达情况。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中TNF-α蛋白表达的WB代表图片;(B)各组TNF-α蛋白WB的灰度值统计结果。
图17是缺血再灌注后HIF-1α免疫荧光结果。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中HIF-1α的荧光染色代表图片。(Scale bar=100μm)。(B)模型组(MCAO)、化合物2组、化合物1组各组脑组织中HIF-1α的阳性细胞密度统计结果。
图18是缺血再灌注后HIF-1α的mRNA表达量情况。假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中HIF-1α的mRNA统计结果。
图19是缺血再灌注后各组HIF-1α蛋白表达情况。(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中HIF-1α蛋白表达的WB代表图片。(B)各组HIF-1α蛋白WB的灰度值统计结果。
具体实施方式
本发明人经过广泛而深入的研究,首次意外发现了具有式I所示结构的化合物,或其药学上可接受的盐、或其溶剂化物、或其前药是一种可有效治疗缺血性脑卒中的活性成分。实验表明,如式I所示的化合物能够抑制星形胶质细胞与小胶质细胞的增生,降低促炎因子(TNF-α、IL-1β、IL-6、iNOS、COX2)及HIF-1α的表达水平,通过抑制炎症的发生发展从而达到治疗缺血性脑卒中的效果。在此基础上,发明人完成了本发明。
治疗缺血性脑卒中的活性成分
在本发明中,提供了一种可以治疗缺血性脑卒中的活性成分。该活性成分为如式(I)所示的化合物,或其药学上可接受的盐、其光学异构体,水合物、溶剂化物或前药;
其中,X选自下组:O或S;
R1和R2各自独立地选自下组:OH、SH、NH2、X2-PO(OH)2、或X2-PS(OH)2
X2选自下组;O或S;
表示
其中,优选的化合物为化合物1或化合物2:
实验表明,本发明的活性成分能够抑制星形胶质细胞与小胶质细胞的增生,降低促炎因子(TNF-α、IL-1β、IL-6、iNOS、COX2)及HIF-1α的表达水平,通过抑制炎症的发生发展从而达到治疗缺血性脑卒中的效果。
如本文所用,“活性成分”、“本发明的活性化合物”、“本发明的活性成分”可互换使用,均指本发明的式I化合物及其结构类似物。
应理解,本发明的活性成分包括本发明的式I化合物、或其药学上可接受的盐、对映异构体、非对映异构体或外消旋体、或其前药。应理解,本发明的活性成分还包括本发明的式I化合物的各种晶型、无定形化合物、以及氘代化合物等形式。
所述“药学上可接受的盐”为式(I)化合物与无机碱形成的钠盐、钾盐、钙盐、铝盐或铵盐;或者式(I)化合物与有机碱形成的甲胺盐、乙胺盐或乙醇胺盐;或者式(I)化合物与赖氨酸、精氨酸、鸟氨酸形成酯后再与盐酸、氢溴酸、氢氟酸、硫酸、硝酸或磷酸形成的对应的无机酸盐或与甲酸、乙酸、苦味酸、甲磺酸或乙磺酸形成的对应的有机酸盐。在本发明中,优选的一类药学上可接受的盐为铵盐,更优选地为五铵盐。
药物组合物和应用
本发明还提供了式I化合物、或其药学上可接受的盐、对映异构体、非对映异构体或外消旋体及前药中的一种或多种的混合物为有效成分在制备治疗和/或缓解缺血性脑卒中等相关疾病的药物中的用途。
本发明所提供的药物组合物优选含有重量比为0.001-99wt%的活性成份,优选的比例是式I化合物作为活性成分占总重量的0.1wt%~90wt%,其余部分为药学可接受的载体、稀释液或溶液或盐溶液。
需要的时候,在本发明药物中还可以加入一种或多种药学上可接受的载体。所述载体包括药学领域常规的稀释剂、赋形剂、填充剂、粘合剂、润湿剂、崩解剂、吸收促进剂、表面活性剂、吸附载体、润滑剂等。
本发明所提供的化合物和药物组合物可以是多种形式,如片剂、胶囊、粉剂、糖浆、溶液状、悬浮液和气雾剂等,并可以存在于适宜的固体或液体的载体或稀释液中和适宜的用于注射或滴注的消毒器具中。
本发明的药物组合物的各种剂型可按照药学领域的常规制备方法制备。其制剂配方的单位计量中通常包含0.05-400mg式(I)化合物,优选地,制剂配方的单位计量中包含1mg-500mg式(I)化合物。
本发明的化合物和药物组合物可对哺乳动物临床使用,包括人和动物,可以通 过口、鼻、皮肤、肺或者胃肠道等的给药途径,最优选为注射制剂(如输液剂)。最优选日剂量为0.01-400mg/kg体重,一次性服用,或0.01-200mg/kg体重分次服用。不管用何种服用方法,个人的最佳剂量应依据具体的治疗而定。通常情况下是从小剂量开始,逐渐增加剂量一直到找到最适合的剂量。
本发明的药物或抑制剂可通过各种不同方式施用,例如可通过注射、喷射、滴鼻、滴眼、渗透、吸收、物理或化学介导的方法导入机体如肌肉、皮内、皮下、静脉、粘膜组织;或是被其他物质混合或包裹导入机体。
本发明优点包括:
(1)本发明的式I化合物可以明显地改善缺血再灌注引起的脑梗死,显著提高缺血再灌注小鼠的运动协调平衡性,明显改善其神经功能损伤,同时还能显著抑制小鼠体重的下降,降低小鼠的死亡率。
(2)本发明实验表明式I化合物能够抑制星形胶质细胞与小胶质细胞的增生,降低促炎因子(TNF-α、IL-1β、IL-6、iNOS、COX2)及HIF-1α的表达水平,通过抑制炎症的发生发展从而达到治疗缺血性脑卒中的效果。
(3)本发明化合物的毒副作用低,成药性好。
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,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。
实验动物
实验以雄性C57BL/6小鼠进行研究和脑组织提取。本实验所用动物均取自中国农业科学院兰州兽医研究所。动物在标准环境下饲养:室温(20±2℃),满足昼夜交替12h。实验动物供应水和标准饲料,在进行实验前会适应3天。所有的动物实验进行时间为上午8点至下午6点。本研究涉及的动物实验严格遵循兰州大学实验动物伦理规范执行(许可证号:JCYXY甘2021-0126)。
化合物1和化合物2按照刘慧云.新型血氧调节剂的设计、合成及活性评价[D].兰州大学,2019.中所述的方法进行制备。
一、缺血性脑卒中模型的构建与化合物2的药效评价
实施例1:大脑中动脉阻塞(MCAO)模型的建立及验证
本实验采用参考Longa等人的方法进行构建小鼠MCAO模型,该模型很好地模拟临床上缺血性脑卒中疾病的发生发展与急性期治疗的过程,可为缺血性卒中的预防与治疗机制研究提供了稳定的应用价值。为评价造模是否成功,检测小鼠 术后第一天的脑区梗死体积,实验结果如图1A和图1B所示,通过2,3,5-三苯四氮唑氯化物(2,3,5-triphenyte-trazoliumchloride,TTC)染色结果显示,术后一天即出现脑区梗死,梗死体积所占比例为45.14±4.48%(P<0.001)。在缺血前、缺血期间和再灌注后采用激光散斑血流仪对不同时间的血流进行了测定,实验结果如图1C和图1D所示,大脑中动脉阻塞30min后,血流指数相对于缺血前下降(138.3±5 vs 52.67±2.3,P<0.001)。1h后拔出线栓再灌注,血流恢复,与缺血期间相比血流指数有显著上升(52.67±2.3 vs 95±5,P<0.001)。同时参照Zea-Longa评分筛选法,对造模24h后的小鼠进行行为评分,实验结果如图1E所示,术后第1天小鼠出现明显的卒中现象,爬行时身体出现明显的倾斜,与假手术相比,具有明显的差异(P<0.001)。综合TTC染色结果、血流情况和小鼠行为,本研究建立的MCAO模型是成功的。n=6,数据以mean±SEM表示,采用one-way ANOVA分析方法进行处理分析,并进行Tukey's HSD检验。*P<0.05,**P<0.01,***P<0.001,*表示与SHAM进行比较(图1B与图1E),*表示与Pre-MCAO进行比较(图1C与图1D);#P<0.05,##P<0.01,###P<0.001,*表示与MCAO-30min进行比较。
实施例2:化合物2对MCAO模型小鼠的神经行为学影响
将体重为23±1g雄性C57/BL小鼠随机分为假手术组(SHAM)、模型组(MCAO)和给药组(50mg/kg、100mg/kg、200mg/kg)各6只。SHAM组的小鼠进行血管分离,但不放线栓,手术前后不予治疗;模型组小鼠构建缺血MCAO模型,手术前后不予治疗;给药组在模型组基础上,给予化合物2治疗。剔除再灌注后神经功能学评分为0分的小鼠和术后未至72h死亡的小鼠。于再灌注后0h、1天和2天给药,给药方法为尾静脉给药,SHAM组和MCAO组给予等体积的生理盐水。
神经功能评分反映MCAO模型小鼠的神经损伤状态,造模后24h,进行神经功能评分(参照Zea-Longa评分筛选法)。1-3分的小鼠即造模成功,将得0分和4分者剔除。评分标准如下表:
Zea-Longa评分筛选法
本实验通过Zea-Longa评分方法对假手组术(SHAM)、模型组(MCAO)、化合物2(50mg/kg)组、化合物2(100mg/kg)组和化合物2(200mg/kg)组进 行评分,主要评分时间点为术前、1天、2天、3天。实验结果如图2,假手术组评分为0,表明小鼠未出现神经功能损伤;与假手术组相比,MCAO组和化合物2组在再灌注24h时,小鼠表现出向一侧打圈的明显现象,表明模型构建成功。与模型组相比,化合物2组在再灌注24h时未见明显差异,而在48h后三个浓度的化合物2组评分都有明显下降,其中化合物2(100mg/kg)组的评分下降最明显(P<0.001)。再灌注72h后,化合物2(100mg/kg)药物浓度时的神经功能评分相较于模型组明显下降(P<0.001)。n=6,数据以mean±SEM,采用two-way ANOVA分析方法进行处理分析,并进行Tukey's HSD检验。*P<0.05,**P<0.01,***P<0.001,*与模型组(MCAO)进行比较。
结果表明,化合物2能有效改善模型小鼠的神经功能缺损,改善缺血再灌注后小鼠协调平衡能力,对修复脑缺血再灌注后的损伤可能存在很好的保护作用。
实施例3:化合物2对MCAO模型小鼠的脑梗死体积影响
通过TTC染色检测实施例2中的假手组术(SHAM)、模型组(MCAO)、化合物2(50mg/kg)组、化合物2(100mg/kg)组和化合物2(200mg/kg)的模型小鼠脑梗死体积。结果如图3所示,模型组小鼠脑组织可见明显的缺血梗死灶。不同浓度的给药组的小鼠的脑梗死体积都不相同,与模型组相比,化合物2(100mg/kg)治疗后模型小鼠的梗死体积明显减小(27.17±5.18%,P<0.001),化合物2(200mg/kg)组的小鼠梗死体积也明显减小(16.19±3.56%,P<0.05)。与化合物2给药浓度为50mg/kg相比,给药浓度为100mg/kg的模型小鼠的脑梗死体积明显较小(26.22±4.68%,P<0.05)。n=6,数据以mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey's HSD检验。*P<0.05,**P<0.01,***P<0.001,表示与模型组(MCAO)进行比较。#P<0.05,##P<0.01,###P<0.001,表示与化合物2 50mg/kg进行比较。
二、本发明化合物治疗缺血性脑卒中模型的机制研究
本发明以化合物1和化合物2为例研究本发明化合物治疗缺血性脑卒中的机制。
分组与给药方法:将体重为23±1g雄性C57/BL小鼠随机分为假手术组(SHAM)、模型组(MCAO)、化合物2组(100mg/kg)和化合物1组(50mg/kg)各10只。SHAM组的小鼠进行血管分离,但不放线栓,手术前后不予治疗;模型组小鼠构建MCAO模型,手术前后不予治疗;给药组在模型组基础上,分别给予化合物2或化合物1治疗。剔除再灌注后神经功能学评分为0分的小鼠和术后未至72h死亡的小鼠。动物行为测试的给药时间为再灌注后0h、1天、2天、3天、4天、5天、6天给药七次,其他实验给药时间为再灌注后0h、1天、2天给药三次。给药方法为尾静脉给药,假手术组和模型组给予等量的生理盐水。
实施例4:本发明化合物对MCAO模型小鼠行为功能的影响
缺血再灌注后,神经元受到严重损伤,突触结构同样遭到严重破坏,这些损伤的级联累加最终会导致行为出现严重缺陷。为了研究缺血再灌注后化合物2和化合物1对行为的影响,通过平衡转棒实验,对小鼠缺血再灌注后动作协调性进行了测试分析。
采用平衡转棒对小鼠的运动协调进行测试。实验分为四组:假手术组(SHAM)、模型组(MCAO)、化合物2组与化合物1组,综合分析化合物2对缺血再灌注后小鼠行为功能恢复的影响。转棒测试能测试小鼠的运动协调能力,实验程序设定为300s内转棒的速度由10rpm逐渐加速至40rpm。记录小鼠在转棒上停留的时间。将小鼠提前3天进行训练。选取可以在转棒上停留300s左右的小鼠进行下一步实验。选取构建模型前小鼠在转棒上停留的时间为术前测试值。将训练过的小鼠进行MCA阻塞60min后进行再灌注。在术后1、3、5、7天分别对SHAM组、MCAO组、化合物2组、化合物1组小鼠进行转棒测试,记录小鼠在转棒停留的时间。
实验结果如图4A所示,与假手术组相比,缺血再灌注后1天、3天、5天、7天MCAO组的转棒持续时间均显著性减少(P<0.001),说明缺血再灌注后小鼠的行为损伤十分严重。再灌注1天到3天,动作协调能力持续下降,而3天后逐渐恢复,但恢复的程度极低。与模型组(MCAO)相比,化合物2组在5天和7天时显著改善了小鼠运动协调的能力(P<0.05),再灌注7天时小鼠的动作协调性明显改善(P<0.001)。与MCAO组相比,化合物1组的小鼠运动协调能力也有提高。化合物2组与化合物1组相比,化合物2组的运动协调能力明显提高(P<0.001)。
改良小鼠神经功能缺损评分(mNSS)表(十四分制)

此外,采用上表所示的改良小鼠神经功能缺损评分(mNSS)的方法对每组小鼠进行神经功能学评分(十四分制)。在术后1、3、5、7天分别对SHAM组、MCAO组、化合物2组、化合物1组小鼠进行神经行为学评分。10到14分的小鼠表明神经系统受损程度为严重。评分为5到9分的小鼠表明神经系统受损程度为中等。1到4分指神经系统受损程度为轻度。最大分值为14分。神经功能损伤评分结果分析如图4B所示,与MCAO组相比,化合物2组从再灌注1天到7天,小鼠的神经功能都有显著改善。化合物2组与化合物1组相比,化合物2组的神经功能明显提高。
同时对缺血再灌注后小鼠的体重变化和生存率做了分析。结果如图4C所示,与假手术组相比,缺血再灌注后的小鼠体重显著性降低。再灌注3天时减轻最严重,之后逐渐恢复,但直到7天仍未恢复到缺血前的体重。与MCAO组相比,再灌注7天时,化合物2抑制了缺血再灌注后小鼠体重的下降。与化合物2组相比,化合物1组的小鼠体重在再灌注7天时显著下降(P<0.05)。生存曲线结果如图4D所示,与MCAO组相比,化合物2显著性改善了缺血再灌注后小鼠的死亡率。n=10,数据以mean±SEM,采用two-way ANOVA分析方法进行处理分析,并进行Tukey's HSD检验。*P<0.05,**P<0.01,***P<0.001,#P<0.05,##P<0.01,###P<0.001;*表示与假手术组(SHAM)进行比较。#表示与化合物2组进行比较。
实施例5:本发明化合物对MCAO模型小鼠中星形胶质细胞的作用
为研究化合物2在缺血再灌注中是否对星形胶质细胞活化增生存在影响,本研究通过免疫荧光实验来观察检测假手术组、模型组、化合物2组和化合物1组的脑组织GFAP的荧光表达,接着通过Western blot检测脑组织中星形胶质细胞的标志物GFAP的表达水平。荧光染色与定量分析结果如图5所示。缺血再灌注72h后,通过免疫荧光染色对脑区的星形胶质细胞GFAP蛋白的活化增生变化情况进行分析。荧光染色结果如图5A所示,MCAO组出现大量的GFAP阳性细胞,而化合物2和化合物1组GFAP阳性细胞数目明显减少。统计结果如图5B所示,MCAO模型组的GFAP阳性细胞数目急剧升高。与MCAO组相比,化合物2给 药三天后,GFAP阳性细胞数目显著下降(195±45 vs 481±67/mm2,P<0.001)。与MCAO组相比,化合物1给药三天后,GFAP阳性细胞数目也显著下降(327±50 vs481±67/mm2,P<0.001)。化合物2组与化合物1组相比,化合物2组的GFAP阳性细胞数目降低更明显(195±45 vs 327±50/mm2,P<0.001)。n=3,数据用mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与MCAO组进行比较,#表示与化合物2组进行比较。
缺血再灌注72h后,利用Western blot对脑组织中星形胶质细胞的标志物GFAP的蛋白表达水平进行检测。假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中GFAP蛋白表达的Western blot(WB)代表图片如图6A所示,定量统计结果如图6B所示。与假手术组相比,MCAO组小鼠脑组织的GFAP蛋白表达量明显升高了91±30%(P<0.001)。与MCAO组相比,化合物2给药三天后,GFAP蛋白表达量明显降低了43±12%(P<0.001)。与MCAO相比,化合物1给药三天后,GFAP蛋白表达量明显降低25±10%(P<0.01)。化合物2组与化合物1组相比,化合物2组的GFAP蛋白表达量明显降低45±10%(P<0.05)。该实验以β-actin作为内参蛋白。n=3,数据用mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与SHAM组进行比较,#表示与化合物2组进行比较。
实施例6:本发明化合物对MCAO模型小鼠中小胶质细胞活化增生的影响
小胶质细胞是神经系统常驻吞噬细胞,是中枢神经细胞应激和损伤的初级防御系统。脑缺血再灌注后,小胶质细胞在外界损伤刺激后发生活化,释放促炎介质和抗炎介质。iba1(ionized calcium binding adapter molecule1)在中枢神经系统中的小胶质细胞中特异性表达,是一种约17kDa的钙结合蛋白。为探究在缺血再灌注模型中化合物对小胶质细胞活化增生是否存在影响,本研究通过免疫荧光实验检测假手术组(SHAM)、模型组(MCAO)、化合物2组和化合物1组的脑组织iba1的荧光表达情况。接着通过Western blot检测脑组织中小胶质细胞中iba1的表达水平。缺血再灌注72h后,通过免疫荧光对脑区的小胶质细胞蛋白iba1染色,分析其变化情况。荧光染色结果如图7A所示,MCAO组出现大量iba1阳性细胞信号,而化合物2和化合物1减少了iba1阳性细胞信号。定量分析结果如图7B所示,统计结果表明,MCAO模型组的iba1阳性细胞数目的表达急剧升高。与MCAO模型组相比,化合物2给药三天后,iba1阳性细胞数目显著下降(148±40 vs 303±23/mm2,P<0.001)。与MCAO模型组相比,化合物1给药三天后,iba1阳性细胞数目也显著下降(200±12 vs 303±23/mm2,P<0.001)。化合物2组与化合物1组相比,化合物2组的iba1阳性细胞数目更低(148±40 vs 200±12/mm2, P<0.001)。n=3,数据用mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与MCAO组进行比较,#表示与化合物2组进行比较。
通过Western blot对脑组织中小胶质细胞的特异性蛋白iba1的表达水平进行检测,(A)假手术组(SHAM)、模型组(MCAO)、化合物2组、化合物1组各组脑组织中iba1蛋白表达的WB代表图片如图8A所示,定量统计结果如图8B所示。结果表明,与假手术组相比,MCAO模型组小鼠脑组织的iba1蛋白表达量明显升高110±50%(P<0.001)。与MCAO组相比,化合物2给药三天后,iba1蛋白表达量明显降低50±12%(P<0.05)。与MCAO组相比,化合物1给药三天后,iba1蛋白表达量降低。化合物2组与化合物1组相比,化合物2组的iba1蛋白表达量降低。该实验以β-actin作为内参蛋白。n=3,数据用mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与SHAM组进行比较,#表示与化合物2组进行比较。
结果发现,相比于模型组,化合物2组和化合物1组的小胶质细胞和星形胶质细胞表达量均明显下降,但化合物2治疗能够显著抑制缺血后脑区的小胶质细胞和星形胶质细胞的过度激活。
实施例7:本发明化合物对MCAO模型小鼠中炎症因子的表达水平的影响
目前缺血性脑卒中的确切病因尚不清楚,但研究表明炎症反应在缺血性脑卒中发病过程中起着重要作用。炎症因子的表达变化可作为缺血性脑损伤治疗效果的有效评价指标。血源性白细胞向脑实质的浸润和内源性小胶质细胞的激活是脑缺血后引发强烈炎症反应的原因。在缺血条件下,脑组织中小胶质细胞和浸润的免疫细胞显著增加。小胶质细胞通过释放促炎因子(TNF-α、IL-1β和IL-6等)和细胞毒性分子(IFN-γ、前列腺素、ROS和NO等)促进神经炎症。小胶质细胞可能通过如下机制导致神经元损伤:1)分泌促炎因子;2)激活氮氧化物(NOX),导致小胶质细胞增生与神经炎症;3)表达iNOS;4)对神经元的胞吞作用。环氧化酶(cyclooxygenase,COX)在前列腺素生物合成过程中起到关键作用。COX2作为双加氧酶和过氧化物酶,介导花生四烯酸形成前列腺素,在炎症发生过程中起到关键作用。COX2受特定事件刺激后进行诱导表达,如应对感染和炎症等生理应激反应,产生前列腺素。
为探究化合物对缺血再灌注后炎症因子表达的影响,本研究通过免疫荧光实验来观察脑组织TNF-α、IL-1β、IL-6、iNOS和COX2蛋白的表达情况,通过RT-qPCR实验检测假手术组、模型组、化合物2组和化合物1组的促炎因子TNF-α、IL-1β、IL-6、iNOS和COX2的mRNA基因表达情况。通过酶联免疫(ELISA)检测脑组织匀浆液中TNF-α、IL-1β和IL-6的浓度,通过Western blot检测脑组织 中TNF-α蛋白的表达水平。
缺血再灌注72h后,通过免疫荧光染色对脑区的TNF-α、IL-1β、IL-6、iNOS和COX2等炎症因子的表达变化做了分析。结果如图9至图13所示。MCAO组出现大量促炎因子阳性细胞信号,而化合物2和化合物1降低了促炎因子阳性细胞信号。统计结果表明,MCAO组的促炎因子阳性细胞数急剧升高,与MCAO模型组相比,化合物2给药三天后,促炎因子的阳性细胞数目显著下降(P<0.001)。统计结果表明,MCAO模型组的iba1阳性细胞数目急剧升高。与MCAO组相比,化合物1给药三天后,促炎因子阳性细胞数目也有显著下降(P<0.001)。化合物2组与化合物1组相比,化合物2组的促炎因子阳性细胞密度显著降低(P<0.001)。如图9所示,MCAO模型小鼠在给予化合物2治疗后,TNF-α阳性细胞的数目显著下降(140±35 vs 361±48/mm2,P<0.001),化合物1的TNF-α阳性细胞的数目虽有下降(244±36 vs 361±48/mm2,P<0.001),但抑制效果不如化合物2组显著。其他促炎因子IL-1β、IL-6、iNOS和COX2表现出与TNF-α相近的趋势。n=3,数据用mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与MCAO组进行比较,#表示与化合物2组进行比较。
RT-qPCR实验结果与定量分析结果如图14所示,缺血再灌注72h后,通过RT-qPCR对脑组织中TNF-α、IL-1β、IL-6、iNOS和COX2的mRNA表达量进行检测分析。结果表明,与假手术组相比,MCAO组的TNF-α(P<0.001)、IL-1β(P<0.001)、IL-6(P<0.01)、iNOS(P<0.001)和COX2(P<0.001)的表达量有不同程度的升高。与MCAO组相比,化合物2给药三天后,TNF-α(P<0.05)、IL-1β(P<0.01)、IL-6(P<0.01)、iNOS(P<0.05)和COX2(P<0.001)的表达量明显降低。其中化合物2对IL-6的表达量抑制效果最明显,下降了51±8%。与MCAO组相比,化合物1给药三天后,TNF-α(P<0.05)、iNOS(P<0.05)和COX2(P<0.001)的表达量在不同程度上都明显降低。n=3,数据用mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与SHAM组进行比较,#表示与化合物2组进行比较。
ELISA实验结果与定量分析结果如图15所示。缺血再灌注72h后,检测小鼠脑组织匀浆液中的炎症因子TNF-α、IL-1β、IL-6的表达水平。结果表明,与假手术组相比,MCAO组的TNF-α(P<0.01)、IL-1β(P<0.001)、IL-6(P<0.01)的表达量有不同程度的升高。与MCAO组相比,化合物2给药三天后,TNF-α、IL-1β和IL-6的表达量都明显降低,其中TNF-α的表达量减降低40±10%(P<0.01),IL-1β的表达量减降低32±9%(P<0.001),IL-6的表达量降低40±10%(P<0.05)。与MCAO组相比,化合物1给药三天后,TNF-α、IL-1β(P<0.05)、IL-6的表达量在不同程度上降低。n=3,数据用mean±SEM,采用one-way ANOVA分析方法 进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与SHAM组进行比较,#表示与化合物2组进行比较。
缺血再灌注72h后,通过Western blot对脑组织中TNF-α蛋白的表达水平检测。实验与定量分析结果如图16所示,与假手术组相比,MCAO组小鼠脑组织中TNF-α的蛋白表达量明显升高120±50%(P<0.001)。与MCAO组相比,化合物2给药三天后,TNF-α蛋白表达量明显降低40±12%(P<0.001)。与MCAO组相比,化合物1给药三天后,TNF-α蛋白表达量降低20±10%(P<0.001)。化合物2组与化合物1组相比,化合物2组的TNF-α的蛋白表达量降低31±20%(P<0.05)。该实验以β-actin作为内参蛋白。n=3,数据用mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与SHAM组进行比较,#表示与化合物2组进行比较。
本研究中,MCAO模型小鼠脑组织匀浆液中TNF-α、IL-1β和IL-6的浓度明显高于假手术组。经过化合物2治疗后脑组织中TNF-α、IL-1β和IL-6的含量明显下降,化合物1组脑组织匀浆液中TNF-α、IL-1β和IL-6的产生也低于模型组。本研究表明化合物在治疗缺血再灌注损伤时,可能通过抑制炎症因子表达,减轻缺血性脑梗死后的炎症,改善脑缺血带来的损伤,且化合物2的治疗效果比化合物1更加明显。
实施例8:本发明化合物对MCAO模型小鼠中缺氧诱导因子的影响
HIF-1α是一种敏感的氧稳态调节因子,在缺氧缺血后迅速诱导其表达。它在卒中的病理生理学中发挥着广泛的作用,包括神经元存活、神经炎症、血管生成、糖代谢和血脑屏障调节。它不仅可以在机体缺氧的情况下产生特异性感受,还能为机体氧稳态的维持过程提供便利。本研究通过免疫荧光实验来观察脑组织HIF-1α的表达水平,通过Western blot检测脑组织中HIF-1α的表达水平,以及通过RT-qPCR实验检测假手术组、模型组、化合物2组和化合物1组的相关因子基因表达情况。
荧光染色结果与定量分析结果如图17所示。缺血再灌注72h后,通过免疫荧光染色对脑区的炎症因子HIF-1α的表达变化进行分析。染色结果显示,假手术组几乎无HIF-1α阳性的细胞出现,MCAO组有大量HIF-1α阳性细胞出现,而化合物2和化合物1减少了促炎因子阳性细胞数。统计结果表明,与假手术组相比,MCAO模型组的HIF-1α阳性细胞数目急剧升高。与MCAO组相比,化合物2给药三天后,HIF-1α阳性细胞数目显著下降(197±33 vs 568±60/mm2,P<0.001)。与MCAO组相比,化合物1给药三天后,HIF-1α阳性细胞数目也显著下降(250±54 vs 568±60/mm2,P<0.001)。化合物2组与化合物1组相比,化合物2组的HIF-1α阳性细胞数目更低(197±33 vs 250±54/mm2,P<0.001)。n=3,数据用mean±SEM, 采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与MCAO组进行比较,#表示与化合物2组进行比较。
缺血再灌注72h后,通过RT-qPCR对脑组织中HIF-1α的mRNA表达量进行检测分析。RT-qPCR实验与定量分析结果如图18所示,与假手术组相比,MCAO组的HIF-1α的表达量明显升高417±107%(P<0.001)。与MCAO组相比,化合物2给药三天后,HIF-1α的表达水平显著降低75±17%(P<0.001);化合物1给药三天后,HIF-1α的表达量也明显降低41±13%(P<0.01)。化合物2组与化合物1组相比,化合物2组的HIF-1α的表达量降低53±11%(P<0.001)。n=3,数据用mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与SHAM组进行比较,#表示与化合物2组进行比较。
缺血再灌注72h后,通过Western blot对脑组织中HIF-1α蛋白的表达水平检测,实验与定量分析结果如图19所示,与假手术组相比,MCAO模型组小鼠脑组织的HIF-1α的蛋白表达量明显升高50±20%(P<0.001)。与MCAO模型组相比,化合物2给药三天后,HIF-1α蛋白表达量明显降低53±10%(P<0.001)。与MCAO模型组相比,化合物1给药三天后,HIF-1α蛋白表达量降低46±10%(P<0.01)。化合物2组与化合物1组相比,化合物2组的HIF-1α蛋白表达量明显降低。该实验以β-actin作为内参蛋白。n=3,数据用mean±SEM,采用one-way ANOVA分析方法进行处理分析,并进行Tukey’s HSD检验。*P<0.05,**P<0.01,***P<0.001;#P<0.05,##P<0.01,###P<0.001。*表示与SHAM组进行比较,#表示与化合物2组进行比较。
结果表明:相较于假手术组,模型组HIF-1α蛋白表达明显升高;相比模型组,化合物2组和化合物1组的HIF-1α蛋白表达明显降低,且化合物2组较于化合物1组降低更多。本研究通过免疫荧光实验证实化合物2及化合物1能明显降低由于缺血再灌注损伤引起的HIF-1α的表达。此外,通过RT-qPCR实验探讨基因水平HIF-1α的表达情况,结果显示化合物2及化合物1明显降低由于缺血再灌注损伤引起的HIF-1α的上调。综上所述,化合物2及化合物1可能通过调节HIF-1α的表达,触发下游的通路,从而起到治疗缺血再灌注损伤的作用。
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 (10)

  1. 一种如下式I所示的化合物,或其药学上可接受的盐、其光学异构体,水合物、溶剂化物或前药的用途;
    其中,X选自下组:O或S;
    R1和R2各自独立地选自下组:OH、SH、NH2、X2-PO(OH)2、或X2-PS(OH)2
    X2选自下组;O或S;
    表示
    其特征在于,用于制备用于治疗和/或缓解缺血性脑卒中的药物组合物。
  2. 如权利要求1所述的用途,其特征在于,所述的式I化合物具有选自下组的结构:
    其中,X的定义如上文中所述。
  3. 如权利要求1所述的用途,其特征在于,所述的式I化合物具有选自下组的结构:
  4. 如权利要求1所述的用途,其特征在于,所述的式I化合物具有选自下组的结构:
    其中,X的定义如上文中所述。
  5. 如权利要求1所述的用途,其特征在于,所述的药学上可接受的盐选自下组: 碱金属盐、碱土金属盐、铵盐。
  6. 如权利要求1所述的用途,其特征在于,所述的药物组合物还用于抑制星形胶质细胞与小胶质细胞的增生。
  7. 如权利要求1所述的用途,其特征在于,所述的药物组合物用于改善和/或缓解缺血性脑卒中引起的炎症反应。
  8. 如权利要求7所述的用途,其特征在于,所述的药物组合物还用于降低促炎因子的表达水平。
  9. 如权利要求1所述的用途,其特征在于,所述的促炎因子选自下组:TNF-α、IL-1β、IL-6、iNOS或COX2。
  10. 如权利要求1所述的用途,其特征在于,所述的药物组合物还用于降低氧稳态调节因子HIF-1α的表达水平。
PCT/CN2023/092591 2022-05-18 2023-05-06 一种酚酸衍生物用于治疗缺血性脑卒中的应用 WO2023221793A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210599845.1 2022-05-18
CN202210599845.1A CN117122588A (zh) 2022-05-18 2022-05-18 一种酚酸衍生物用于治疗缺血性脑卒中的应用

Publications (1)

Publication Number Publication Date
WO2023221793A1 true WO2023221793A1 (zh) 2023-11-23

Family

ID=88834541

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/092591 WO2023221793A1 (zh) 2022-05-18 2023-05-06 一种酚酸衍生物用于治疗缺血性脑卒中的应用

Country Status (2)

Country Link
CN (1) CN117122588A (zh)
WO (1) WO2023221793A1 (zh)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050222096A1 (en) * 2002-08-29 2005-10-06 Greenfield Robert S Methods of screening for compounds that modulate TAFIa activity, compounds, and methods of using the compounds
CN114191425A (zh) * 2021-12-13 2022-03-18 亚宝药业集团股份有限公司 2,4-二羟基苯甲酸在治疗铁过载疾病食品药品中的应用

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050222096A1 (en) * 2002-08-29 2005-10-06 Greenfield Robert S Methods of screening for compounds that modulate TAFIa activity, compounds, and methods of using the compounds
CN114191425A (zh) * 2021-12-13 2022-03-18 亚宝药业集团股份有限公司 2,4-二羟基苯甲酸在治疗铁过载疾病食品药品中的应用

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ABEDI FARSHAD, RAZAVI BIBI MARJAN, HOSSEINZADEH HOSSEIN: "A review on gentisic acid as a plant derived phenolic acid and metabolite of aspirin: Comprehensive pharmacology, toxicology, and some pharmaceutical aspects", PHYSIOTHERAPY RESEARCH, JOHN WILEY & SONS LTD. CHICHESTER., GB, vol. 34, no. 4, 1 April 2020 (2020-04-01), GB , pages 729 - 741, XP093108119, ISSN: 0951-418X, DOI: 10.1002/ptr.6573 *
XIAOLI JIAN, YIN SIYUAN, ZHANG DECHOU: "Advances in the study of immune cell changes after ischemic stroke", JOURNAL OF SOUTHWEST MEDICAL UNIVERSITY, vol. 43, no. 5, 20 October 2020 (2020-10-20), pages 524 - 527, XP093108158, DOI: 10.3969/j.issn.2096-3351.2020.05.020 *

Also Published As

Publication number Publication date
CN117122588A (zh) 2023-11-28

Similar Documents

Publication Publication Date Title
JP3020808B2 (ja) 精神病治療薬としての4−フェニル−1,2,3,4−テトラヒドロ−1−ナフタレンアミン誘導体の使用
US20090048288A1 (en) Method of treating stress-mediated depression
EA021303B1 (ru) Способы лечения или профилактики рвоты с помощью агентов, усиливающих секрецию гормона роста
Kapin et al. Neuroprotective effects of eliprodil in retinal excitotoxicity and ischemia.
TW201505634A (zh) 藥物戒斷之無毒治療方法
EP2589382A1 (en) Pharmaceutical composition comprising levocarnitine and dobesilate
US11524055B2 (en) Methods for treating diseases mediated by ERBB4-positive pro-inflammatory macrophages
CA2957224A1 (en) Method of treating prader-willi syndrome
WO2023221793A1 (zh) 一种酚酸衍生物用于治疗缺血性脑卒中的应用
US20110118317A1 (en) Use of epothilones in the treatment of neuronal connectivity defects such as schizophrenia and autism
JPH07505608A (ja) インターロイキン−1阻害化合物による神経病学的状態の治療
WO2023202102A1 (zh) 倍半萜类化合物在抑制trpa1通道的活性中的应用
JPH07508258A (ja) モノシアロガングリオシドgm↓1またはその誘導体を含有する,パーキンソン病の治療に適する医薬組成物
JP2005511639A (ja) 虚血性疾患の治療用ドパミン再取り込み阻害活性を有するトロパン誘導体
US20180200231A1 (en) Il-8 inihibitors for use in the treatment of some urological disorders
CN113117090A (zh) 以胞内蛋白纳米颗粒渗透压为靶点治疗人体水肿的药物组合物及应用
RU2721282C2 (ru) Способ лечения рассеянного склероза (варианты)
US20020058706A1 (en) Method for treating asthma using pregabalin
WO2022183493A1 (zh) 一种多酚类化合物的应用
JP6796056B2 (ja) 自閉症の治療
JP7257091B2 (ja) 認知症の治療及び予防薬
US11730769B2 (en) Compositions and methods for Williams Syndrome (WS) therapy
CN112043700B (zh) 盐酸去亚甲基四氢小檗碱在制备预防或治疗神经退行性疾病药物中的应用
US11446264B2 (en) Memory manipulation via modification of protein kinase C zeta activity
WO2024061251A1 (zh) 一种4-tmap治疗或缓解抑郁症的用途

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: 23806745

Country of ref document: EP

Kind code of ref document: A1