CN111358793B - Novel application of HUWE1 inhibitor BI8622 - Google Patents

Novel application of HUWE1 inhibitor BI8622 Download PDF

Info

Publication number
CN111358793B
CN111358793B CN202010176486.XA CN202010176486A CN111358793B CN 111358793 B CN111358793 B CN 111358793B CN 202010176486 A CN202010176486 A CN 202010176486A CN 111358793 B CN111358793 B CN 111358793B
Authority
CN
China
Prior art keywords
inflammasome
cells
activation
huwe1
inhibitor
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
CN202010176486.XA
Other languages
Chinese (zh)
Other versions
CN111358793A (en
Inventor
齐晓朋
徐涛
李艺辉
郭晓敏
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.)
Kunming Institute of Zoology of CAS
Original Assignee
Kunming Institute of Zoology of CAS
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 Kunming Institute of Zoology of CAS filed Critical Kunming Institute of Zoology of CAS
Priority to CN202010176486.XA priority Critical patent/CN111358793B/en
Priority to PCT/CN2020/085013 priority patent/WO2021179396A1/en
Publication of CN111358793A publication Critical patent/CN111358793A/en
Application granted granted Critical
Publication of CN111358793B publication Critical patent/CN111358793B/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
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Diabetes (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Epidemiology (AREA)
  • Emergency Medicine (AREA)
  • Dermatology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Endocrinology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention provides a new application of HUWE1 inhibitor BI8622 in inhibiting activation of inflammasome, and researches show that: the HUWE1 inhibitor BI8622 can remarkably reduce the activation of cysteine protease-1 (Caspase-1) and the expression of interleukin 1 beta (IL-1 beta) in mouse bone marrow-derived macrophages (BMDM), and reduce the occurrence of cell death events; meanwhile, the BI8622 is proved to be capable of remarkably reducing the activation of Caspase-1 and the expression of IL-1 beta and remarkably reducing the cell death in a human monocyte/macrophage line THP1 and human primary Peripheral Blood Mononuclear Cells (PBMC), so that the BI8622 can remarkably inhibit the activation of inflammatory corpuscles.

Description

Novel application of HUWE1 inhibitor BI8622
Technical Field
The invention relates to the technical field of medicines, and in particular relates to a new application of HUWE1 inhibitor BI 8622.
Background
Activation of the inflammasome plays an important role in the development of inflammation, resistance to pathogen invasion, and development of autoimmune diseases in the host. Classical inflammatory bodies include NLRP3, AIM2 and NLRC4 inflammatory bodies, which are activated by recognition of different stimuli, respectively, and ultimately all result in the activation of cellular cysteine protease-1 (Caspase-1), thereby inducing the production of mature interleukin 1 beta (IL-1 beta), interleukin 18(IL-18), activated Caspase-1 cleavable gut dermatan D (GSDMD), leading to the occurrence of cellular apoptosis (Inflamamosomes: mechanism of action, role in disease, and thermoeutics Haitao Guo, Justin B Callaway & Jenny P-Y Ting; Nature medicinal volume 21, pages 677-687 (2015)). Abnormal activation of inflammasome is associated with the development of a variety of diseases, and more drugs are developed by targeting activation of inflammasome.
HUWE1 inhibitor BI8622 is a class of small molecule compounds having the following structural formula:
Figure BDA0002410998720000011
BI8622 is obtained by screening by Stefanie Peter et al, can specifically inhibit ubiquitin ligase HUWE1 through ubiquitination modification function, and finds that the inhibitor has a certain inhibiting effect on tumorigenesis and development (Peter S, Bumultick J, Myant K, Jaenicke LA, Walz S, Muller J, et al. Tumor cell-specific inhibition of MYC functional using small molecular inhibitors of the HUWE1ubiquitin ligand. EMBO Mol. Med. 2014; 6: 5-.
Disclosure of Invention
The invention aims to solve the technical problem of providing an application of HUWE1 inhibitor BI8622 in preparing a medicine for inhibiting activation of inflammatory corpuscles.
The chemical structural formula of BI8622 of the invention is as follows:
Figure BDA0002410998720000021
in the present invention, the inflammasome includes NLRP3, AIM2 and NLRC 4.
In the invention, the NLRP3 inflammasome is activated by lipopolysaccharide and adenosine triphosphate together, the AIM2 inflammasome is activated by Francisella or dsDNA, the NLRC4 inflammasome is activated by Salmonella, and the NLRP3 and AIM2 inflammasome are simultaneously activated by Listeria.
The invention provides a medicament for inhibiting activation of an inflammasome, which comprises an active ingredient BI8622 and a pharmaceutically acceptable carrier.
The invention provides application of HUWE1 inhibitor BI8622 in preparation of medicines for treating gout, type II diabetes mellitus or inflammatory diseases of systemic lupus erythematosus.
The invention provides a medicament for treating inflammatory diseases such as gout, type II diabetes mellitus or systemic lupus erythematosus, which comprises an active ingredient BI8622 and a pharmaceutically acceptable carrier.
Compared with the prior art, the technical scheme of the invention has the following beneficial effects:
according to the invention, the BI8622 has effects on activated NLRP3, AIM2 and NLRC4 inflammatory corpuscles and cell death on mouse primary BMDM cells, human monocyte macrophage line THP1 cells and human primary PBMC cells respectively, and the results show that the BI8622 can remarkably inhibit the activation of the inflammatory corpuscles and remarkably reduce the occurrence of cell death events in the mouse BMDM cells, the human monocyte macrophage line THP1 cells and the human primary PBMC cells.
The invention proves the new application of HUWE1 inhibitor BI8622 for the first time: can be used for inhibiting and verifying activation of corpuscles, and shows that BI8622 has potential medicinal value in treating inflammatory corpuscle abnormal activation diseases (such as gout, II type diabetes, systemic lupus erythematosus and other inflammatory diseases).
Drawings
FIG. 1 is a graph comparing the expression of activated caspase 1P20 in mouse primary BMDM cells after treatment by BI8622 treatment and control;
FIG. 2 is a graph comparing IL-1 β expression levels in mouse primary BMDM cells after treatment with BI8622 treatment and control;
FIG. 3 is a graph comparing TNF expression levels in mouse primary BMDM cells after treatment with BI8622 and a control;
FIG. 4 is a graph comparing IL-6 expression levels in mouse primary BMDM cells after treatment with BI8622 treatment and control;
FIG. 5 is a graph comparing the rate of cell death in primary mouse BMDM after treatment with BI8622 and a control;
FIG. 6 is a graph comparing the IL-1. beta. expression levels of human THP1 cells after treatment with BI8622 and a control group;
FIG. 7 is a graph comparing TNF expression levels in human THP1 cells after treatment with BI8622 and a control;
FIG. 8 is a graph comparing the IL-6 expression levels of human THP1 cells after treatment with BI8622 and a control group;
FIG. 9 is a graph comparing the rate of human THP1 cell death after treatment with BI8622 and a control;
FIG. 10 is a graph comparing the expression levels of IL-1 β in human primary PBMC cells after treatment with BI8622 and a control group;
FIG. 11 is a graph comparing TNF expression levels in human primary PBMC cells after treatment with BI8622 and a control group;
FIG. 12 is a graph comparing the expression levels of IL-6 in human primary PBMC cells after treatment with BI8622 and a control group;
FIG. 13 is a graph comparing the proportion of human primary PBMC cell death following treatment with BI8622 and control.
Detailed Description
In a specific embodiment of the present invention, healthy C57/BL mice were obtained from kunming animal research institute of chinese academy of sciences [ experimental animal license number: SYXK (Dian) K2013-0013 ].
The HUWE1 inhibitor BI8622 of the present invention was purchased from MedChemexpress, Inc. under the designation HY-120929.
The human monocyte-macrophage line THP1 used in the present invention is derived from Kunming animal research institute cell bank, Chinese academy of sciences, and other monocyte-macrophage cells in the art can also be used.
NLRP3 inflammasome is typically activated by exogenous molecules such as gram negative bacterial cell wall components, nucleic acids, perforin, endogenous molecules such as Adenosine Triphosphate (ATP), uric acid crystals, etc. (case s.l., Eisenbarth s.c., i yer s.s., et al, The nap 3 fluorescence is an infection for The development of silicon, proc.natl.acad.sci.usa,2008,105(26):9035-9040. kannegatroni t.d., Body-Malapel m, Amer a., 2006, critical roll for Cryopropyrin/nap 3 in activation of pase-1 response to viral infection and 19. biological chemical rna.281 j.281, 3648).
The NLRC4 inflammasome is activated by Salmonella (Salmonella) infection (Mariatasan S., Newton K., Monack D.M., et al. differential activation of the inflamasome by caspase-1adapt ASC and Ipaf. Nature,2004,430(6996): 213-218.).
AIM2 inflammasome is activated by Francisella and double-stranded DNA (dsDNA) induction (Fernandes-Alnemri T., Yu J. -W., Juliana C., et al, The AIM2 inflamasome is criterial for in not natured to Francisella tularensis. Nat. Immunol.2010,11:385-393. Brckst. hammer T., Baumann C., Blu ml S., et al, an organic proteinaceous-genetic materials AIM2 a cytoplasmic DNA sensor for The in not natrumame. Immunol.2009,10: 272).
In the present invention, all the raw material components are commercially available products well known to those skilled in the art unless otherwise specified.
The technical solution of the present invention will be clearly and completely described below with reference to the embodiments of the present invention. It is to be understood that the described embodiments are merely exemplary of the invention, and not restrictive of the full scope of the invention. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
Example 1
BI8622 was tested for the effect of three activated inflammasome and cell death in mouse primary BMDM cells NLRP3, AIM2, NLRC 4.
The material and the method are as follows:
preparation of primary mouse BMDM cells: euthanizing adult (e.g. 4-6 week old) C57/BL mice, separating the femur and tibia under sterile conditions and placing them in PBS buffer, scraping off the muscle, cartilage and epiphysis on the bone surface with a scalpel, cutting the bone to expose the medullary cavity, flushing the medullary cavity with 1mL DMEM-F12 medium aspirated by a 1mL syringe, collecting cells, uniformly blowing the cells, counting and adjusting the cell density to 1-2Million/mL, adding 20ng/mL M-CSF growth factor to the DMEM-F12 complete medium, plating the cells on a cell culture plate, inoculating the cells in 5% CO 5%237 ℃ CO2Culturing in an incubator, and collecting adherent cells for corresponding experiments after 5 days of static culture.
The test compound BI8622 is prepared into a stock solution with the concentration of 10mM by using dimethyl sulfoxide (DMSO), the cell is diluted to a working concentration of 10uM by using DMEM-F12 complete culture medium before being treated, a control group is treated by using 0.1% DMSO, the treatment time of the test group BI8622 and the control group is 3 hours, and then subsequent inflammatory corpuscle stimulant stimulation and detection experiments are carried out.
The inflammasome stimulators are: 1) lipopolysaccharide (LPS) + Adenosine Triphosphate (ATP) that activates NLRP3 inflammasome; 2) francisella (f. novicida) and double-stranded dna (dsdna) that activate AIM2 inflammasome; 3) salmonella (Salmonella) which activates NLRC4 inflammasome; 4) listeria (Listeria) can activate both NLRP3 and AIM2 inflammasome.
The specific stimulation mode is as follows: stimulating with LPS with a final concentration of 500ng/mL for 1h and ATP with a final concentration of 5mM for 15min, thereby activating NLRP3 inflammasome; activating AIM2 inflammasome by stimulating dsDNA transfected with 1.5ug X-fect for 1h or f.novicida at 200MOI for 12 h; stimulating with Salmonella at a final concentration of 3MOI for 2h, thereby activating NLRC4 inflammasome; stimulation with Listeria at a final concentration of 30MOI for 3h activated NLRP3 and AIM2 inflammasome simultaneously.
Meanwhile, culture supernatant (medium) without added stimulus was selected as a 0h control group.
Detecting the expression level of a cysteine protease-1 (Caspase-1) activated cleavage product P20 in culture supernatant and the expression level of a Caspase-1 precursor in cell lysate by using western blot; simultaneously detecting the expression levels of interleukin 1 beta (IL-1 beta), Tumor Necrosis Factor (TNF) and interleukin 6(IL-6) by using an enzyme-linked immunosorbent assay (ELISA); cell death detection (LDH method) was performed by measuring the level of Lactate Dehydrogenase (LDH) secretion in the culture supernatant.
The results showed that in mouse primary BMDM cells, BI8622 treated group cells showed significantly reduced expression of activated caspase-1P20 after LPS + ATP, dsDNA, f. novicida, Salmonella, and Listeria stimulation treatments compared to DMSO control group cells (fig. 1, where Med group represents culture supernatant control without added stimulus and caspase1 represents caspase-1). ELISA test results show that after LPS + ATP, F.novicida, Salmonella and Listeria stimulation treatment, IL-1 beta secretion of cells in BI8622 treated group is obviously lower than that of cells in control group, IL-1 beta expression level of cells in dsDNA treated group is lower, and no difference exists between two groups of cells (FIG. 2); the expression level of TNF was not significantly different in both groups of cells in the above treatment regimes (FIG. 3); IL-6 expression was not significantly different in cells from the other stimulation treated groups except that BI8622 treated group was reduced compared to the control group after f.novicida, Salmonella stimulation (fig. 4). LDH assay results indicated that cell death was lower in BI8622 treated group than in DMSO control group cells in all of the above stimulation regimens (FIG. 5). The above results show that BI8622 can significantly inhibit the activation of NLRP3, AIM2 and NLRC4 inflammasome in mouse BMDM cells, and reduce cell death.
Example 2
BI8622 was tested for its effect on three inflammasome and cell death activated in human THP1 cells, NLRP3, AIM2, NLRC 4.
The THP1 cell is human monocyte macrophage, the culture condition is that 1640 culture medium is added with 10% FBS, and the specific culture method is according to the conventional method. The test compound BI8622 is prepared into a stock solution with the concentration of 10mM by using dimethyl sulfoxide (DMSO), the stock solution is diluted into a working concentration of 10uM by using 1640 complete culture medium before treating cells, a control group is treated by using 0.1% DMSO, the treatment time of the test group BI8622 and the control group is 3 hours, and then subsequent inflammatory corpuscle stimulant stimulation and detection experiments are carried out.
The inflammasome stimulators are: 1) lipopolysaccharide (LPS) + Adenosine Triphosphate (ATP) that activates NLRP3 inflammasome; 2) double stranded dna (dsdna) that activates AIM2 inflammasome; 3) salmonella (Salmonella) which activates the NLRC4 inflammasome.
The specific stimulation mode is as follows: stimulating with LPS with a final concentration of 500ng/mL for 1h and ATP with a final concentration of 5mM for 15min, thereby activating NLRP3 inflammasome; the dsDNA transfection was stimulated for 1h with a final concentration of 1.5ug X-fect, activating AIM2 inflammasome; stimulation with Salmonella at a final concentration of 3MOI for 2h activated NLRC4 inflammasome.
Meanwhile, culture supernatant (medium) without added stimulus was selected as a 0h control group.
ELISA method is used for detecting the expression of IL-1 beta, TNF and IL-6, and LDH method is used for detecting the cell death.
The results showed that after THP1 cells were treated with LPS + ATP, dsDNA, Salmonella stimulation, BI 8622-treated cells secreted significantly less IL-1 β than control cells (fig. 6); BI8622 treated cells also showed lower expression levels of TNF and IL-6 than control cells (FIG. 7); the expression level of TNF in the cells did not differ significantly in any of the above-described treatment modalities (fig. 8); LDH assay also revealed that BI 8622-treated THP1 cells died significantly less after LPS + ATP, dsDNA, Salmonella treatment than did control cells (FIG. 9). The results show that, similar to mouse BMDM cells, BI8622 can remarkably inhibit the activation of NLRP3, AIM2 and NLRC4 inflammasome in human THP1 cells, and can obviously reduce the death of the cells.
Example 3
BI8622 was tested for the effect of three inflammasome and cell death activated in human primary PBMC cells NLRP3, AIM2, NLRC 4.
Peripheral Blood Mononuclear Cells (PBMCs) were obtained by density gradient centrifugation of 10mL of peripheral blood taken from normal donors [ approved by the ethical committee of life science of kunming institute of science and technology, code: SMKX-20191201-01 ]. The PBMC cells obtained by separation are cultured in 1640 medium + 10% FBS, and the specific culture method can be according to the conventional method. The test compound BI8622 is prepared into a stock solution with the concentration of 10mM by using dimethyl sulfoxide (DMSO), the stock solution is diluted into a working concentration of 10uM by using 1640 complete culture medium before treating cells, a control group is treated by using 0.1% DMSO, the treatment time of the test group BI8622 and the control group is 3 hours, and then subsequent inflammatory corpuscle stimulant stimulation and detection experiments are carried out. The stimulus and the specific stimulation mode were specifically selected in example 2, and the culture supernatant (medium) without the stimulus was used as a 0h control group. Expression of IL 1-beta, TNF and IL-6 was measured by ELISA and cell death was measured by LDH.
The results showed that after PBMC cells were stimulated with LPS + ATP, dsDNA, Salmonella, the secretion of IL-1. beta. was significantly lower in BI 8622-treated cells than in control cells (FIG. 10); expression of TNF in cells of BI8622 treated group was lower than control PBMC cells after LPS + ATP and Salmonella stimulation, there was no significant difference in TNF expression in cells of both groups stimulated by dsDNA (fig. 11); IL-6 expression levels in PBMCs from BI8622 treated groups were significantly lower after Salmonella stimulation than in control groups, but not significantly different after LPS + ATP and dsDNA stimulation (FIG. 12). LDH detection also found that cellular LDH secretion levels were significantly lower in the BI 8622-treated human primary PBMCs after LPS + ATP, dsDNA, Salmonella treatment than in the control PBMCs, suggesting that the BI 8622-treated human primary PBMCs died significantly lower than in the control (fig. 13). The above results show that BI8622 can significantly inhibit the activation of NLRP3, AIM2 and NLRC4 inflammasome in human primary PBMC, and can significantly reduce cell death.
From the results in the examples, the present invention demonstrates for the first time the novel use of HUWE1 inhibitor BI 8622: BI8622 can both significantly inhibit the activation of inflammatory bodies and significantly reduce the occurrence of cell death events.
The foregoing is only a preferred embodiment of the present invention, and it should be noted that, for those skilled in the art, various modifications and decorations can be made without departing from the principle of the present invention, and these modifications and decorations should also be regarded as the protection scope of the present invention.

Claims (3)

  1. Use of an HUWE1 inhibitor BI8622 for the manufacture of a medicament for the treatment of inflammatory diseases such as gout, type II diabetes, or systemic lupus erythematosus, wherein the HUWE1 inhibitor BI8622 inhibits the activation of inflammasome, said inflammasome being NLRP3, AIM2, and NLRC 4.
  2. 2. The use of claim 1, wherein said BI8622 has the following chemical structure:
    Figure FDA0002944064770000011
  3. 3. the use of claim 1, wherein said NLRP3 inflammasome is activated by both lipopolysaccharide and adenosine triphosphate, said AIM2 inflammasome is activated by francisella or dsDNA, said NLRC4 inflammasome is activated by salmonella, and said NLRP3 and AIM2 inflammasome are simultaneously activated by listeria.
CN202010176486.XA 2020-03-13 2020-03-13 Novel application of HUWE1 inhibitor BI8622 Active CN111358793B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202010176486.XA CN111358793B (en) 2020-03-13 2020-03-13 Novel application of HUWE1 inhibitor BI8622
PCT/CN2020/085013 WO2021179396A1 (en) 2020-03-13 2020-04-16 New application of huwe1 inhibitor bi8622

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010176486.XA CN111358793B (en) 2020-03-13 2020-03-13 Novel application of HUWE1 inhibitor BI8622

Publications (2)

Publication Number Publication Date
CN111358793A CN111358793A (en) 2020-07-03
CN111358793B true CN111358793B (en) 2021-08-10

Family

ID=71198563

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010176486.XA Active CN111358793B (en) 2020-03-13 2020-03-13 Novel application of HUWE1 inhibitor BI8622

Country Status (2)

Country Link
CN (1) CN111358793B (en)
WO (1) WO2021179396A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113244237B (en) * 2021-06-22 2022-03-04 南京市儿童医院 Application of BI8622 in preparation of medicine for relieving acute kidney injury induced by cisplatin
CN116236563A (en) * 2023-01-12 2023-06-09 佛山病原微生物研究院 Application of ASB17 in preparation of medicine for preventing or treating inflammation caused by salmonella typhimurium

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104140954A (en) * 2013-05-09 2014-11-12 北京大学 Method for regulating expression level of HUWE1 in cells
CN108938697A (en) * 2018-09-21 2018-12-07 南华大学 Arasaponin is preparing the application in NLRP3 inflammation corpusculum inhibition drug or medicament for treatment of depression
CN110251657B (en) * 2019-06-14 2020-11-17 中山大学 Application of EBV BRLF1 and functional small peptide thereof in inhibiting activity of inflammatory corpuscles
CN110227075A (en) * 2019-07-01 2019-09-13 中国人民解放军总医院第五医学中心 Purposes of the carnosol as inflammation corpusculum inhibitor

Also Published As

Publication number Publication date
CN111358793A (en) 2020-07-03
WO2021179396A1 (en) 2021-09-16

Similar Documents

Publication Publication Date Title
CN111437279B (en) Application of HUWE1 inhibitor BI8626 in preparation of medicines for inhibiting activation of inflammasome
Lei et al. Interleukin-17 induces pyroptosis in osteoblasts through the NLRP3 inflammasome pathway in vitro
Ortiz et al. Experimental microbial dysbiosis does not promote disease progression in SIV-infected macaques
CN111358793B (en) Novel application of HUWE1 inhibitor BI8622
Castelli et al. Exosome secretion by Leishmania infantum modulate the chemotactic behavior and cytokinic expression creating an environment permissive for early infection
Zhang et al. Rosmarinic acid protects mice from imiquimod induced psoriasis‐like skin lesions by inhibiting the IL‐23/Th17 axis via regulating Jak2/Stat3 signaling pathway
Navarro-Partida et al. Pirfenidone restricts Th2 differentiation in vitro and limits Th2 response in experimental liver fibrosis
Chen et al. Elp3‐mediated codon‐dependent translation promotes mTORC2 activation and regulates macrophage polarization
JP2021167345A (en) Modulating inflammasome activation of myeloid-derived suppressor cells for treating gvhd or tumor
Yang et al. The increased expression of an array of CXC and CC chemokines in the colonic mucosa of patients with ulcerative colitis: regulation by corticosteroids
Zheng et al. Establishing transcription profile of psoriasiform cutaneous in vitro using HaCaT cells stimulated with combination of cytokines
Chen et al. A paracrine circuit of IL-1β/IL-1R1 between myeloid and tumor cells drives glioblastoma progression
Gonzalez Suarez et al. EGCG inhibits the inflammation and senescence inducing properties of MDA-MB-231 triple-negative breast cancer (TNBC) cells-derived extracellular vesicles in human adipose-derived mesenchymal stem cells
Sun et al. Rebalancing liver-infiltrating CCR3+ and CD206+ monocytes improves diet-induced NAFLD
Rojas et al. Serotype a of Aggregatibacter actinomycetemcomitans down-regulates the increased serotype b-induced cytokine and chemokine production in dendritic cells
Li et al. Matrine alleviates imiquimod-induced psoriasiform dermatitis in BALB/c mice via dendritic cell regulation
Zhang et al. Granulocyte macrophage colony stimulating factor is elevated in alveolar macrophages from sheep naturally infected with maedi-visna virus and stimulates maedi-visna virus replication in macrophages in vitro
Fadhil et al. Evaluation of some biochemical markers in relation to COVID-19 mRNA vaccine
Usui et al. Thymus-expressed chemokine enhances Porphyromonas gingivalis LPS-induced osteoclast formation via NFATc1 activation
Wu et al. A monocyte-keratinocyte-derived co-culture assay accurately identifies efficacies of BET inhibitors as therapeutic candidates for psoriasiform dermatitis
Shusha Unique Cellular Senescence Phenotype and Inflammatory Response in Cells Cultured from Long-Lived Fruit Bats
Xiao et al. β-Glucan augments IL-1β production by activating the JAK2/STAT3 pathway in cultured rabbit keratinocytes
Rasoulpoor et al. A mimic of the tumor microenvironment on GPR30 gene expression in breast cancer
CN110585228B (en) Application of LncRNA PTPRE-AS1 in preparation of medicine for inhibiting allergic asthma induced by cockroach extract
Liang et al. Viperin inhibits interferon-γ production to promote Mycobacteria tuberculosis infection by disrupting TBK1-IKKε-IRF3-axis and JAK-STAT signaling

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