CN113813387B - Application of PPAR agonist in preparing medicine for treating acute myeloid leukemia - Google Patents

Application of PPAR agonist in preparing medicine for treating acute myeloid leukemia Download PDF

Info

Publication number
CN113813387B
CN113813387B CN202111239178.8A CN202111239178A CN113813387B CN 113813387 B CN113813387 B CN 113813387B CN 202111239178 A CN202111239178 A CN 202111239178A CN 113813387 B CN113813387 B CN 113813387B
Authority
CN
China
Prior art keywords
ppar
aml
sitagliptin
sodium
alpha
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202111239178.8A
Other languages
Chinese (zh)
Other versions
CN113813387A (en
Inventor
徐兵
周辉
鲁先平
潘德思
李志峰
查洁
付鑫
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chengdu Weixin Pharmaceutical Co ltd
Shenzhen Chipscreen Biosciences Co Ltd
First Affiliated Hospital of Xiamen University
Original Assignee
Chengdu Weixin Pharmaceutical Co ltd
Shenzhen Chipscreen Biosciences Co Ltd
First Affiliated Hospital of Xiamen University
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 Chengdu Weixin Pharmaceutical Co ltd, Shenzhen Chipscreen Biosciences Co Ltd, First Affiliated Hospital of Xiamen University filed Critical Chengdu Weixin Pharmaceutical Co ltd
Priority to CN202111239178.8A priority Critical patent/CN113813387B/en
Publication of CN113813387A publication Critical patent/CN113813387A/en
Priority to PCT/CN2022/127184 priority patent/WO2023072010A1/en
Priority to TW111140518A priority patent/TWI835370B/en
Application granted granted Critical
Publication of CN113813387B publication Critical patent/CN113813387B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • C12N5/0694Cells of blood, e.g. leukemia cells, myeloma cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/385Hormones with nuclear receptors of the family of the retinoic acid recptor, e.g. RAR, RXR; Peroxisome proliferator-activated receptor [PPAR]

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biomedical Technology (AREA)
  • Oncology (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides application of a PPAR agonist in preparing a medicament for treating acute myelocytic leukemia. Through an AML cell strain and a CDX model, the invention researches and obtains the action mechanism of the PPAR agonist for inhibiting the generation and the development of AML as follows: PPAR agonist represented by sitagliptin sodium is combined with transcription factor PPAR alpha, so as to inhibit ubiquitination degradation of PPAR alpha and activate protein expression of PPAR alpha in AML cells; after PPAR alpha is activated, the expression of hypoxia inducible factor HIF1 alpha and downstream target genes PGK1, glut1, LDHA, PGM, MCT4 and HK2 thereof is further reduced, the glycolysis process of AML cells is inhibited, and finally the generation and development process of AML is inhibited. Therefore, the PPAR agonist is expected to become a medicine with wide application prospect for treating acute myeloid leukemia.

Description

Application of PPAR agonist in preparing medicine for treating acute myelocytic leukemia
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to an application of a PPAR agonist in preparation of a medicine for treating acute myeloid leukemia.
Background
Acute Myeloid Leukemia (AML) is a malignant clonal hematopoietic disease affecting hematopoietic stem and progenitor cells, generally caused by abnormal genetic or epigenetic changes that increase the likelihood of cell self-renewal and malignant proliferation, and also mutations in some key genes that contribute to myeloid differentiation in the mid-AML progression (see f.a. lagunas-range, v.ch a vez-Valencia, M).
Figure BDA0003318690660000011
Log of mez-Guijosa, C.cortex-Penagos, inlet myelid Leucoemia-genetic disorders and the ir clinical prognosis, int.J.Hematol.Oncol.Stem Cell Res.11 (4) (2017) 328-339), the progression of AML involves oncogene mutations, epigenetic modifications and metabolic disorders. The median age of AML patients is 68 years, and in recent years the incidence of AML has steadily risen annually and the prognosis is poor.
Traditional therapeutic options for AML are limited to high-dose cytotoxic chemotherapy, and with the molecular-based studies on AML, more and more therapeutic targets are discovered, and molecular targeted therapies have superior efficacy and lower toxicity compared to traditional chemotherapy. Since 2017, the U.S. food and drug administration approved at least 8 new drugs for treating AML in different settings, including small molecule inhibitors, antibody drug conjugates, and cytotoxic drugs; the common small molecule inhibitor sorafenib (sorafenib) is a multikinase inhibitor, active on a variety of receptor tyrosine kinases, including VEGF, ras, raf and FLT3, with superior efficacy in FLT3-ITD mutated AML (see Ravandi F, alattar ML, grunwald MR, et al, phase 2study of azacytidine plus sorafenib in tissues with ingredient muscle tissue free and FLT-3internal mutation therapy, blood, 2013jun 6 (121) (23): 4655-4662), although patients develop resistance or are refractory to treatment.
Several studies have shown that tumor cells are finely dependent on glucose uptake and utilization, sugar metabolism plays an important role in tumor cell growth, and that enhanced glycolysis has been shown by Chen et al to contribute to the reduction of sensitivity of the anti-AML drug Ara-c, while inhibition of glycolysis can further inhibit AML cell proliferation and enhance the cytotoxicity of Ara-c (see Chen WL, wang JH, zhao AH, xu X, wang YH, chen TL, li JM, miq, zhu YM, liu YF, wang YY, chen Z, chen SJ, jia w. Additive glucose metabolism signature of ingredient myoid blood with blood vessel Sep 2014 4. 2014 124 (10 1645-54.Doi 10.2/blood-2014-5502-4204. Jub 2014, 2014 18. Meyer 2014 18.
The Xiglitazar sodium (Chiglitazar) is a diabetes treatment drug which is designed and synthesized by Shenzhen micro-core biotechnology, inc. in China and has brand-new chemical structure and global intellectual property protection, has PPAR alpha, PPAR delta and PPAR gamma full activation, belongs to a new generation of insulin sensitizer and is mainly used for treating type 2 diabetes (see Cheng HS, tan WR, low ZS, marvalim C, lee JYH, tan NS.amplification and Development of PPAR Modulators in Health and Disease: an Update of Clinical evaluation. Int J Mol Sci.2019 Oct 11 (20): 5055).
However, the mechanism of action of PPAR agonists in AML therapy and AML-associated cell lines is unknown.
Disclosure of Invention
Aiming at the defects in the prior art, the invention aims to provide the application of a PPAR agonist in preparing a medicament for treating acute myeloid leukemia, in particular to the application of the PPAR agonist with the PPAR alpha agonist activity in preparing a medicament for preventing and/or treating the acute myeloid leukemia.
Besides the PPAR agonist can be combined with the PPAR alpha to activate the PPAR alpha and inhibit the ubiquitination degradation of the PPAR alpha, the PPAR agonist is proved to be capable of inhibiting the glycolysis process of an AML cell strain at a molecular level and inhibiting the proliferation and inducing the apoptosis level of the AML cell strain at a cell level, and the PPAR agonist represented by the sitagliptin sodium is also proved to be capable of inhibiting the tumor formation process of the AML of a mouse through a CDX mouse model in an animal experiment.
To achieve the purpose, the invention provides the following technical scheme:
in a first aspect, the present invention provides the use of a PPAR agonist in the manufacture of a medicament for the prevention and/or treatment of acute myeloid leukaemia.
In the invention, an AML cell strain and a CDX model prove that a PPAR agonist, in particular to sitagliptat sodium (Chiglitazar), can inhibit the proliferation of AML cells and induce the apoptosis of the AML cells, can also inhibit the tumor formation process of NSG mouse AML and improve the survival rate, in the aspect of the model, the sitagliptat sodium can be combined with PPAR alpha and inhibit the ubiquitination degradation of the PPAR alpha, so as to activate the protein expression of the PPAR alpha in the AML cells, and further down-regulate the expressions of hypoxia inducible factor HIF1 alpha and downstream target genes PGK1, glut1, LDHA, PGM, MCT4 and HK2 thereof after the PPAR alpha is activated, thereby inhibiting the glycolysis process of the AML cells and finally inhibiting the generation and development process of the AML.
The invention also provides application of the PPAR agonist in preventing and/or treating acute myeloid leukemia.
In some embodiments, the PPAR agonist has PPAR α agonist activity.
In some embodiments, the PPAR agonist is selected from one or a combination of at least two of the following drugs: ciguatazoxy (Chiglitazar), fenofibrate (Fenofibrate), gemfibrozil (Gemfibrozil), fenofibric Acid (Fenofibric Acid), flufenamic Acid (Flufenamic Acid), ibuprofen (Ibuprofen), bezafibrate (Bezafibrate), indomethacin (Indomethacin), rosiglitazone (Rosglitazone), ciprofibrate (Ciprofibrate), valproic Acid (Valproic Acid), dexibuprofen (Dexibuprofen), amiodarone (Amidolone), prasterone (Prasterone), alpha-evening primrose oil Acid (alpha-linear enacid), PPM-204 (Indolitexazar), clinoflurate (Clinoflor), myrrh (Myrrh), palmitic Acid (Paitmix), fenoprofen (Acnovafin), lauric Acid (Laurac), ibuprofen (Clinoflor), ibuprofen (Ibuprofen), ibuprofen (Clinoflurandr), ibuprofen (Paxib), and the like Stearic Acid (Stearic Acid), clofibrate (Clofibrate), docosahexaenoic Acid (Doconexent), oleic Acid (Oleic Acid), troglitazone (Troglitazone), omega-3fatty acids (Omega-3 fatty acids), eicosapentaenoic Acid (Icosapent), myristic Acid (Myristic Acid), arachidonic Acid (arachidic Acid), isoflavone (isoflazone), azaglitazar (Aleglitazar), regoraza (regoraza), GFT505 (Elafibrandor), migalaza (Muragitazar), rositaloxaza (Ertiprostafib), phthalic Acid (Phthalic Acid), laggeraza (Ragaglitazar), tesaglitazar (Tesagalzar), leukotrine B4 (Lerietone B4), leuconabenza (Leucona 4), leucona-5935, and Leucona-D4, octanoic acid (Caprylic acid), GW501516 (Cardarine), resveratrol (Resveratrol), N-Bis (3- (D-glucamide) propyl) deoxycholamide (N, N-Bis (3- (D-gluconamidodo) propyl) deoxycholestyramide) or isomers, solvates, metabolites, crystalline forms, amorphous forms or pharmaceutically acceptable salts thereof.
In some embodiments, the PPAR agonist is selected from any one of the group consisting of cyglifloxacarb or an isomer, solvate, metabolite, crystalline form, amorphous form, or a pharmaceutically acceptable salt thereof.
In some embodiments, the pharmaceutically acceptable salt is any one of an alkali metal salt, an alkaline earth metal salt, an ammonium salt or a quaternary ammonium salt, preferably an alkali metal salt, and more preferably a sodium salt and a potassium salt.
In some embodiments, the isomer is the levorotatory form.
In some embodiments, the PPAR agonist is sitagliptin sodium, sitagliptin potassium, or an levorotatory form thereof.
In a second aspect, the present invention provides a pharmaceutical composition for preventing and/or treating acute myeloid leukemia, which comprises a PPAR agonist.
In some embodiments, the pharmaceutical composition further comprises a pharmaceutically acceptable excipient.
In some embodiments, the pharmaceutically acceptable excipients include any one of or a combination of at least two of a carrier, diluent, excipient, filler, binder, wetting agent, disintegrant, emulsifier, cosolvent, solubilizer, tonicity modifier, surfactant, coating material, colorant, pH adjuster, antioxidant, bacteriostatic or buffer.
In some embodiments, the dosage form of the pharmaceutical composition includes any one of tablets, powders, suspensions, granules, capsules, injections, sprays, solutions, enemas, emulsions, films, suppositories, patches, nasal drops or dropping pills.
In some embodiments, the pharmaceutical composition has a PPAR agonist as the only active ingredient.
In some embodiments, the pharmaceutical composition may include other active ingredients in addition to the PPAR agonist. The other active ingredients may act synergistically with the PPAR agonist or they may act additively with the PPAR agonist, preferably without antagonism of the PPAR agonist.
In a third aspect, the present invention also provides the use of a PPAR agonist in the preparation of an AML cell line proliferation inhibitor, an AML cell line apoptosis promoter, or an AML cell line cell cycle blocker.
The invention also provides the application of the PPAR agonist in inhibiting the proliferation of AML cell strains, promoting the apoptosis of AML cell strains or retarding the cell cycle of AML cell strains.
In some embodiments, the AML cell line is an acute myeloid leukemia-inducing cell line.
In some embodiments, the AML cell line comprises KG-1 alpha cells and/or Kasumi cells.
In the invention, experiments prove that the PPAR agonist can inhibit the proliferation of AML cell strains and induce the apoptosis level of the AML cell strains by taking the sitagliptin sodium as a research object.
In a fourth aspect, the invention also provides application of the PPAR agonist in preparing a medicament for inhibiting the glycolysis of AML cells, down-regulating the hypoxia inducible factor HIF1 alpha or reducing the expression level of glycolysis related genes.
The invention also provides application of the PPAR agonist in inhibiting the glycolysis of AML cells, down-regulating the hypoxia inducible factor HIF1 alpha or reducing the expression level of glycolysis related genes.
In some embodiments, the glycolytic related gene includes any one of PGK1, MCT4, glut1, PGM, LDHA, or HK2 or a combination of at least two thereof.
In the invention, western blot, RT-PCR and glucose consumption detection prove that the PPAR agonist can inhibit the glycolysis process of an AML cell line by taking the sodium sitagliptin as a research object.
In addition, the PPAR agonist sitagliptin sodium is proved to be capable of inhibiting the tumorigenic process of mouse AML by a CDX mouse model in the invention.
In a fifth aspect, the present invention provides a method of preventing and/or treating acute myeloid leukemia, said method comprising administering to a subject a prophylactically and/or therapeutically effective amount of said PPAR agonist.
Compared with the prior art, the invention has the beneficial effects that:
according to the invention, the inhibition effect of a PPAR agonist, particularly a PPAR alpha type agonist, on acute myelocytic leukemia and a molecular mechanism thereof are mainly explored by taking the sitagliptin sodium as a research object, and the inhibition effect of the PPAR agonist, particularly the PPAR alpha type agonist, on the acute myelocytic leukemia and the molecular mechanism thereof are firstly proved according to an Autodock molecular docking modeling and a Co-IP experiment that the sitagliptin sodium can be combined with a transcription factor PPAR alpha and can inhibit ubiquitination degradation of the PPAR alpha so as to activate protein expression of the PPAR alpha in AML cells; meanwhile, the sodium sitagliptin is proved to be capable of inhibiting the proliferation of AML cell strains and inducing the apoptosis of the AML cell strains at a cell level; western blot, RT-PCR, glucose consumption and lactose generation detection prove that the sitagliptin sodium can inhibit the glycolysis process of AML cell strains; the CDX mouse model proves that the sitagliptin can inhibit the tumorigenic process of mouse AML; according to experimental results, after activating PPAR alpha, the expression of hypoxia inducible factor HIF1 alpha and downstream target genes PGK1, glut1, LDHA, PGM, MCT4 and HK2 of the hypoxia inducible factor are further reduced, the glycolysis process of AML cells is inhibited, and finally the AML generation and development process is inhibited; according to the above experimental results, PPAR agonists represented by cetostertal sodium can be used as an effective drug for the treatment of acute myeloid leukemia.
Drawings
FIG. 1A is a graph showing the results of the inhibition of cell proliferation of KG-1. Alpha. Cells treated with sodium sitagliptin in example 1 for 48h and 72 h.
FIG. 1B is a graph showing the results of the inhibition of cell proliferation of Kasumi cells treated with sodium sitagliptin in example 1 for 48h and 72 h.
FIG. 2A is a graph showing the results of horizontal flow assay of apoptosis of KG-1 α cells treated with sitagliptin sodium in example 2 after 48h and 72 h.
FIG. 2B is a graph showing the statistics of the apoptosis rate of KG-1. Alpha. Cells in example 2.
FIG. 2C is a graph showing the results of horizontal flow assay of apoptosis in Kasumi cells treated with sodium sitagliptin in example 2 for 48h and 72 h.
FIG. 2D is a graph showing statistics of the apoptosis rate of Kasumi cells in example 2.
FIG. 3A is a graph showing the statistics of the percentage of cells in the cell cycle at G1, S and G2 stages after treatment of Kasumi cells with sitagliptin sodium in example 3.
FIG. 3B is a graph of the statistics of the percentage of cells in the G1, S and G2 phases of the cell cycle after treatment of KG-1 α cells with sitagliptin sodium in example 3.
FIG. 4A is a schematic diagram showing a molecular docking model of sitagliptin sodium with PPAR α in example 4.
FIG. 4B is a graph showing the results of Western blot analysis of KG-1 α cells treated with different concentrations of sodium sitagliptat in example 4 for 48 h.
FIG. 4C is a graph showing the results of Western blot analysis of Kasumi cells treated with different concentrations of sodium sitagliptin in example 4 for 48 hours.
FIG. 5A is a Western blot result of the expression levels of hypoxia inducible factor H1F1 alpha and glycolysis related genes LDHA and PGK1 after KG-1 alpha cells are treated for 48H with different concentrations of Semagita sodium in example 5.
FIG. 5B is a statistical graph of mRNA levels of the glycolysis-related genes PGK1, MCT4, glut1, PGM, LDHA and HK2 after 48h treatment of KG-1 α cells with various concentrations of sitagliptin in example 5.
Fig. 5C is a statistical graph of the results of measuring glucose consumption levels 48h after treatment of KG-1 α cells with different concentrations of sitagliptin sodium in example 5.
FIG. 5D is a histogram of the results of measuring lactose production levels 48h after treatment of KG-1 α cells with different concentrations of cilastatin sodium in example 5.
FIG. 5E is a graph showing the Western blot results of the expression levels of hypoxia inducible factor H1F1 alpha and glycolysis-related genes LDHA and PGK1 after treating Kasumi cells for 48H with different concentrations of SelagrittNa in example 5.
FIG. 5F is a statistical graph of mRNA levels of glycolysis-related genes PGK1, MCT4, glut1, PGM, LDHA and HK2 after 48h treatment of Kasumi cells with different concentrations of sitagliptin sodium in example 5.
FIG. 5G is a histogram showing the results of measuring the glucose consumption levels of Kasumi cells treated with different concentrations of sodium sitagliptat in example 5 for 48 hours.
FIG. 5H is a histogram showing the results of measuring the lactose production levels 48H after treatment of Kasumi cells with different concentrations of sodium sitagliptin in example 5.
Fig. 6A is a graph of in vivo images at day 0, day 7 and day 14 after injection of sitagliptin sodium into the CDX mouse model in example 6.
Fig. 6B is a statistical graph of survival rates of mice after injection of sitagliptin sodium into the CDX mouse model in example 6.
FIG. 7 is a schematic diagram showing the mechanism of action of sitagliptin sodium in inhibiting AML development.
Detailed Description
The technical solutions of the present invention are further described in the following embodiments with reference to the drawings, but the following examples are only simple examples of the present invention and do not represent or limit the scope of the present invention, which is defined by the claims.
In the following examples, reagents and consumables used were obtained from conventional reagent manufacturers in the field unless otherwise specified; unless otherwise indicated, all experimental methods and technical means are conventional in the art.
Example 1 inhibition of proliferation levels of AML cell lines by sitagliptin sodium
Take 1X 10 4 Respectively inoculating AML cell strains KG-1 alpha and Kasumi in logarithmic phase into a 96-well cell culture plate, wherein the KG-1 alpha and Kasumi cell strains are provided by the institute of hematology of medical college of Xiamen university;
the control group treated the cells with DMSO, and the experimental group was set to 5 μ M, 10 μ M, 15 μ M, 20 μ M, and 25 μ M for the sitagliptin sodium concentration gradient;
the cell proliferation level was measured with CCK8 kit after 48h and 72h treatment, respectively.
The results of the cell proliferation levels obtained are shown in FIG. 1A and FIG. 1B;
IC50 values of sitagliptin sodium measured after KG-1 alpha and Kasumi cells were treated with sitagliptin sodium for 48h and 72h, respectively, are shown in Table 1 below:
TABLE 1
Figure BDA0003318690660000091
In combination with the above results, it was found that sitagliptin can inhibit the proliferation level of AML cells and shows time-and-concentration dependence; the inhibition rate gradually increased with the increase of treatment concentration and treatment time.
Example 2 apoptosis of AML cell lines induced by sitagliptin sodium
Taking 1X 10 5 Inoculating AML cell strains KG-1 alpha and Kasumi in logarithmic phase into a 24-hole cell culture plate respectively;
the control group treated the cells with DMSO, and the gradient concentrations of sitagliptin sodium in the experimental group were set to 5 μ M, 10 μ M, 15 μ M, 20 μ M and 25 μ M;
after 48h and 72h of treatment, the apoptosis level is detected by an Annexin V/PI flow staining method, and the apoptosis rate is counted.
FIGS. 2A and 2B are the horizontal flow assay of apoptosis and the statistical results of apoptosis rate after 48h and 72h of KG-1 α cells treated with different concentrations of sitagliptin sodium;
FIGS. 2C and 2D are horizontal flow assay results of apoptosis and apoptosis rate statistics after 48h and 72h of Kasumi cells treated with different concentrations of sitagliptin sodium;
from the above results, it was found that sitagliptin induces apoptosis of AML cells, increases the level of apoptosis of AML cells, and exhibits concentration dependence.
Example 3 cell cycle arrest of AML cell lines by sitagliptin sodium
Take 1X 10 5 AML cell lines KG-1 alpha and Kasumi respectivelyInoculating into 24-well cell culture plate;
control groups treated cells with DMSO, cetrartat sodium concentration 10 μ M;
after 48h of treatment, cells are collected by centrifugation for 5min at 300g, washed once by PBS, fixed for 4h at 4 ℃ by 75% alcohol, stained for 15min by PI, and then the retardation of the cell cycle is analyzed by flow detection.
The results are shown in fig. 3A and 3B, which show that the numbers of cells in S phase and G2 phase are significantly increased and the number of cells in G1 phase is significantly decreased compared to the control group after AML cell lines are treated with sitagliptin sodium, indicating that the sitagliptin sodium can arrest the progression of AML cell cycle.
Example 4 Effect of sitagliptin on PPAR α ubiquitination levels
Firstly, a docking model of the sitagliptin sodium and the PPAR alpha molecule is established.
The sitagliptin sodium molecular structure model is established by using ChemDraw software, the PPAR alpha structure is from a PDB database, the PDB ID is 3KDT, the docking model is established by using AUTODOCK software and is further modified by using Pymol software;
the resulting model is shown in FIG. 4A, which shows that two amino acid residues LYS-266 and ALA-256 of PPAR α are linked to a sitagliptin sodium molecule; indicating that there is an interaction between sitagliptin sodium and the PPAR α molecule.
Second, the effect of sitagliptin sodium on PPAR α ubiquitination (ubiquitin) levels was investigated.
Take 1X 10 7 Respectively inoculating AML cell strains KG-1 alpha and Kasumi in logarithmic phase into a 10cm cell culture dish;
the control group treated the cells with DMSO, and the experimental group was set at 10 μ M and 20 μ M for sitagliptin sodium (and labeled as Chi 10 μ M and Chi 20 μ M);
after 48h of treatment, cells were collected by centrifugation at 300G for 5min, and after adding 500. Mu.L of RIPA lysate and incubating for 30min, protein A/G beads and Anti-PPAR α antibodies were added, and incubation was performed overnight at 4 ℃ to carry out CO-immunoprecipitation (CO-IP), and further, as a result of Western blot analysis, primary antibodies were used with Anti-PPAR α and Anti-Ubiquitylation antibodies, respectively.
The obtained results are shown in fig. 4B and 4C, PPAR α expression is up-regulated after KG-1 α cells and Kasumi cells are treated with different concentrations of sitagliptin sodium for 48 hours, and the CO-IP experimental results show that the ubiquitination degradation level of PPAR α is obviously inhibited.
The results indicate that sitagliptin sodium binds to transcription factor PPAR α and can inhibit ubiquitination degradation level of PPAR α, thereby activating protein expression of PPAR α.
Example 5 Effect of sitagliptin on the expression of HIF 1. Alpha. And downstream target genes of AML cell line
Take 1X 10 6 Respectively inoculating AML cell strains KG-1 alpha and Kasumi in logarithmic phase into a 6cm cell culture dish;
control groups treated cells with DMSO at concentrations of sitagliptin 10. Mu.M and 20. Mu.M, respectively;
treating for 48h, centrifuging for 5min at 300g, collecting cells, collecting half of the cells, performing ice lysis with 200 μ L RIPA lysate for 1h, extracting total protein, performing Western blot, and detecting protein level expression of HIF1 α and downstream target gene thereof;
adding 500 mu L Trizol reagent into the other half cell, extracting by using an RNA extraction kit to obtain total RNA, further performing a reverse transcription experiment by using a reverse transcription kit to obtain total cDNA, and finally designing a primer to perform RT-PCR to detect the expression condition of HIF1 alpha and downstream target genes thereof at the mRNA level.
Cell supernatants were collected and used to measure glucose consumption and lactose production levels by glucose assay kits and lactose production assay kits.
The results of KG-1 α cells are shown in fig. 5A, 5B, 5C and 5D, and Western blot results of protein expression levels after treatment with different concentrations of sitagliptin sodium show that the expression levels of HIF1 α and its downstream target genes LDHA and PGK1 are significantly reduced; meanwhile, reverse transcription experiments show that the mRNA level expression of glycolysis related genes PGK1, MCT4, glut1, PGM, LDHA and HK2 is obviously reduced; finally, the glucose consumption and the lactose generation level are obviously reduced by detecting the glucose and the lactose.
Similarly, the results obtained by Kasumi cells are shown in FIG. 5E, FIG. 5F, FIG. 5G and FIG. 5H, and the Western blot results of the protein expression levels after treatment with different concentrations of Selagita sodium show that the expression levels of HIF1 α and its downstream target genes LDHA and PGK1 are significantly reduced; meanwhile, reverse transcription experiments show that the mRNA level expression of glycolysis related genes PGK1, MCT4, glut1, PGM, LDHA and HK2 is obviously reduced; the consumption of glucose and the production of lactose are significantly reduced.
Example 6 sitagliptin inhibits the tumorigenic process of AML cell transplantable tumors
Stably transfecting a PLV-luciferase-GFP plasmid into a KG-1 alpha cell strain by a lentivirus method to construct an AML cell strain stably expressing luciferase, wherein the PLV-luciferase-GFP plasmid is provided by the institute of hematology of medical college of Xiamen university. Collecting 3X 10 6 Injecting the AML cell strain luciferase-GFP-KG-1 alpha in logarithmic growth phase into NOD-Prkdc through tail vein -/- IL2rg -/- (NSG) mice were tumorigenic, wherein NSG mice were purchased from the university of Xiamen laboratory animal center and were raised by the laboratory animal center.
The tumor formation progress of the mice is detected by a living body imaging system, and the total fluorescence intensity of the mice reaches 1 multiplied by 10 6 -1×10 7 Thereafter (about 14 days), 14days was administered at a dose of 15mg/kg/day, and the day of initiation of administration was taken as 0 day, and the results of the experiment and the survival rate of the mice were counted.
The obtained results are shown in fig. 6A and 6B, and the sitagliptin sodium can obviously inhibit the tumorigenesis process of the CDX model, and the survival rate curve shows that the sitagliptin sodium can obviously improve the survival rate of the CDX model.
The mechanism of action of sitagliptin in inhibiting the development of AML can be analyzed and summarized in combination with the above examples, as shown in fig. 7:
PPAR agonists represented by sitagliptin sodium can be combined with transcription factor PPAR alpha and inhibit ubiquitination degradation of PPAR alpha, so as to activate protein expression of PPAR alpha in AML cells, and further regulate expression of hypoxia inducible factor HIF1 alpha and downstream target genes PGK1, glut1, LDHA, PGM, MCT4 and HK2 thereof after PPAR alpha activation, thereby inhibiting glycolysis process of AML cells and finally inhibiting AML development process.
The applicant declares that the above description is only a specific embodiment of the present invention, but the scope of the present invention is not limited thereto, and it should be understood by those skilled in the art that any changes or substitutions that can be easily conceived by those skilled in the art within the technical scope of the present invention disclosed herein fall within the scope and disclosure of the present invention.

Claims (7)

  1. The application of PPAR agonist in preparing medicine for preventing and/or treating acute myelocytic leukemia;
    the PPAR agonist is selected from any one of the cyglifloxate or levorotatory form, crystal form, amorphous form or pharmaceutically acceptable salt thereof.
  2. 2. The use according to claim 1, wherein the pharmaceutically acceptable salt is any one of an alkali metal salt, an alkaline earth metal salt, an ammonium salt or a quaternary ammonium salt.
  3. 3. The use according to claim 2, wherein the pharmaceutically acceptable salt is an alkali metal salt.
  4. 4. The use according to claim 3, wherein the pharmaceutically acceptable salts are sodium and potassium salts.
  5. 5. The use according to claim 1, wherein the PPAR agonist is sitagliptin sodium, sitagliptin potassium or an levorotatory form thereof.
  6. 6. The use according to claim 1, wherein the PPAR agonist inhibits glycolysis of AML cells, down-regulates the hypoxia inducible factor HIF1 α or decreases the expression level of glycolysis-related genes.
  7. 7. The use of claim 6, wherein the glycolytic related gene comprises any one of PGK1, MCT4, glut1, PGM, LDHA or HK2 or a combination of at least two thereof.
CN202111239178.8A 2021-10-25 2021-10-25 Application of PPAR agonist in preparing medicine for treating acute myeloid leukemia Active CN113813387B (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202111239178.8A CN113813387B (en) 2021-10-25 2021-10-25 Application of PPAR agonist in preparing medicine for treating acute myeloid leukemia
PCT/CN2022/127184 WO2023072010A1 (en) 2021-10-25 2022-10-25 Use of ppar agonist in preparation of drug for treating acute myeloid leukemia
TW111140518A TWI835370B (en) 2021-10-25 2022-10-25 Application of ppar agonist in preparation of medicament for treating acute myeloid leukemia

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111239178.8A CN113813387B (en) 2021-10-25 2021-10-25 Application of PPAR agonist in preparing medicine for treating acute myeloid leukemia

Publications (2)

Publication Number Publication Date
CN113813387A CN113813387A (en) 2021-12-21
CN113813387B true CN113813387B (en) 2023-04-14

Family

ID=78918932

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111239178.8A Active CN113813387B (en) 2021-10-25 2021-10-25 Application of PPAR agonist in preparing medicine for treating acute myeloid leukemia

Country Status (3)

Country Link
CN (1) CN113813387B (en)
TW (1) TWI835370B (en)
WO (1) WO2023072010A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113813387B (en) * 2021-10-25 2023-04-14 厦门大学附属第一医院 Application of PPAR agonist in preparing medicine for treating acute myeloid leukemia
CN114949230A (en) * 2022-06-13 2022-08-30 厦门大学附属第一医院 Combined pharmaceutical composition for preventing and/or treating acute myeloid leukemia and application thereof
CN115068618A (en) * 2022-07-11 2022-09-20 厦门大学附属第一医院 Combined pharmaceutical composition for preventing and/or treating acute myeloid leukemia and application thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111437393B (en) * 2020-04-30 2021-03-02 深圳微芯生物科技股份有限公司 Combined medicine for treating diabetes and complications thereof and pharmaceutical composition thereof
CN111759838B (en) * 2020-07-02 2022-01-25 深圳微芯生物科技股份有限公司 Pharmaceutical composition of pharmaceutically acceptable salt of sitagliptin and application thereof
CN113813387B (en) * 2021-10-25 2023-04-14 厦门大学附属第一医院 Application of PPAR agonist in preparing medicine for treating acute myeloid leukemia

Also Published As

Publication number Publication date
WO2023072010A1 (en) 2023-05-04
TWI835370B (en) 2024-03-11
TW202327580A (en) 2023-07-16
CN113813387A (en) 2021-12-21

Similar Documents

Publication Publication Date Title
CN113813387B (en) Application of PPAR agonist in preparing medicine for treating acute myeloid leukemia
Chen et al. Nrf2 deficiency aggravates Angiotensin II-induced cardiac injury by increasing hypertrophy and enhancing IL-6/STAT3-dependent inflammation
Hu et al. The roles of GRP81 as a metabolic sensor and inflammatory mediator
Liu et al. Emodin alleviates CCl4‑induced liver fibrosis by suppressing epithelial‑mesenchymal transition and transforming growth factor‑β1 in rats
Lu et al. Iminostilbene, a novel small-molecule modulator of PKM2, suppresses macrophage inflammation in myocardial ischemia–reperfusion injury
Farrow et al. NFAT and IRF proteins regulate transcription of the anti-HIV gene, APOBEC3G
Fan et al. Peptidylarginine deiminase IV promotes the development of chemoresistance through inducing autophagy in hepatocellular carcinoma
Wang et al. Effect of Berberine on PPARα/NO Activation in High Glucose‐and Insulin‐Induced Cardiomyocyte Hypertrophy
Li et al. Dragon's Blood exerts cardio-protection against myocardial injury through PI3K-AKT-mTOR signaling pathway in acute myocardial infarction mice model
Lin et al. Qianliening capsule treats benign prostatic hyperplasia via suppression of the EGF/STAT3 signaling pathway
Jiang et al. Ligustilide inhibits the proliferation of non-small cell lung cancer via glycolytic metabolism
Lan et al. Polydatin attenuates hepatic stellate cell proliferation and liver fibrosis by suppressing sphingosine kinase 1
Bai et al. The small molecule P7C3-A20 exerts neuroprotective effects in a hypoxic–ischemic encephalopathy model via activation of PI3K/AKT/GSK3β signaling
Kan et al. Effect of Palrnatine on lipopolysaccharide-induced acute lung injury by inhibiting activation of the Akt/NF-κB pathway
Zhang et al. Identification of a novel PAK1/HDAC6 dual inhibitor ZMF-23 that triggers tubulin-stathmin regulated cell death in triple negative breast cancer
Xu et al. Kukoamine A activates Akt/GSK-3β signaling pathway to inhibit oxidative stress and relieve myocardial ischemia-reperfusion injury
Li et al. Inhibition of poly (ADP-ribose) polymerase inhibits ischemia/reperfusion induced neurodegeneration in retina via suppression of endoplasmic reticulum stress
Sayas Tau-based therapies for Alzheimer’s disease: Promising novel neuroprotective approaches
TW202034919A (en) Use of chiauranib in treating small cell lung cancer
US20210106552A1 (en) Methods for treating triple negative breast cancer using salvianolic acid b
WO2022095976A1 (en) Use of small molecule sr9009 in anti-aging and alleviation of chronic inflammation caused by aging
CN115068618A (en) Combined pharmaceutical composition for preventing and/or treating acute myeloid leukemia and application thereof
Lin et al. Metformin ameliorates neuronal necroptosis after intracerebral hemorrhage by activating AMPK
Koike et al. Pretreatment with olprinone hydrochloride, a phosphodiesterase III inhibitor, attenuates lipopolysaccharide-induced lung injury via an anti-inflammatory effect
Shi et al. MEF2D participates in microglia-mediated neuroprotection in cerebral ischemia-reperfusion rats

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant