Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Br J Cancer ; 127(6): 1014-1025, 2022 10.
Article in English | MEDLINE | ID: mdl-35715638

ABSTRACT

BACKGROUND: Fibroblast growth factor receptor (FGFR) signaling influenced tumour occurrence and development. Overexpression of FGFR had been observed in many types of cancers, including colon cancer. FGFR inhibitor is considered to be effective in treating colon cancer patients. METHODS: First, the kinase inhibition rate was determined. MTT, western blotting, colony formation, EdU and comet assays were performed to evaluate the anti-tumour effects of F1-7 in vitro. RNA-seq and bioinformatics analysis were used for further verification. Additionally, a xenograft model was generated to investigate the anti-tumour effect of F1-7. RESULTS: F1-7 can inhibit the proliferation of colon cancer cells in vitro. It could significantly inhibit FGFR phosphorylation and its downstream signaling pathway. Whole-genome RNA-seq analysis found that the changed genes were not only functionally focused on MAPK signaling pathway but also related to cell apoptosis and ferroptosis. Experimental evidence demonstrated that F1-7 can directly increase the level of cellular DNA damage. The occurrence of DNA damage led to cell cycle arrest and inhibition of cell metastasis and cell apoptosis. Mouse model experiments also confirmed that F1-7 could inhibit tumour growth by inhibiting the FGFR pathway. CONCLUSIONS: F1-7 exhibits anti-tumour activity by inhibiting the FGFR pathway. It could be a novel therapeutic agent for targeting colon cancer cells.


Subject(s)
Colonic Neoplasms , Protein Kinase Inhibitors , Animals , Cell Death , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , DNA Damage , Humans , Mice , Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/genetics
2.
Planta Med ; 88(7): 507-517, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34116570

ABSTRACT

Herbacetin is a bioactive flavanol compound that has various pharmacological effects. However, the pharmacokinetic characteristics have not been thoroughly investigated. Previously, we screened a natural compound library and identified herbacetin as a potent CYP blocker. Herein, we aimed to mechanistically determine the inhibitory effects of herbacetin on CYP450 and its potential application. A human liver microsome incubation system was developed based on a UPLC-MS/MS method. Moreover, an in silico docking assay and a human CYP recombinase reaction system were developed and used to investigate binding affinity and inhibitory efficacy. Subsequently, the effects of the combination of herbacetin and sorafenib on HepG2 cells were assessed by MTT and immunoblotting assays. The concentration of sorafenib and its main metabolite were measured by UPLC-MS/MS after incubation with or without herbacetin. As a result, we found herbacetin almost completely inhibited the functions of major CYPs at 100 µM. Moreover, through analysis of the structure-activity relationship, we found 4-, 6-, and 8-hydroxyl were essential groups for the inhibitory effects. Herbacetin inhibited CYP3A4, CYP2B6, CYP2C9, and CYP2E1 in a mixed manner, but non-competitively blocked CYP2D6. These results are in good agreement with the recombinase reaction in vitro results, with an IC50 < 10 µM for each tested isoenzyme. Interestingly, the stimulatory effects of sorafenib on HepG2 cell apoptosis were significantly enhanced by combining with herbacetin, which was associated with increased sorafenib exposure. In summary, herbacetin is a potent inhibitor of a wide spectrum of CYP450s, which may enhance the exposure of drugs in vivo.


Subject(s)
Microsomes, Liver , Tandem Mass Spectrometry , Chromatography, Liquid , Cytochrome P-450 Enzyme System/metabolism , Flavonoids , Microsomes, Liver/metabolism , Recombinases/pharmacology , Sorafenib/pharmacology
3.
Chem Res Toxicol ; 33(1): 172-180, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31560202

ABSTRACT

As a new atypical antipsychotic, brexpiprazole is primarily metabolized by cytochrome P450 3A4 (CYP3A4). However, genetic polymorphisms in CYP3A4 cause wide variability in individuals' responses to brexpiprazole, leading to unpredictable adverse side effects or even therapeutic failure. The present study was designed to systematically study the effects of 26 recombinant CYP3A4 variants on the metabolism of brexpiprazole and investigate their enzymatic activity. Wild-type CYP3A4 and the 26 variants were incubated with the substrate brexpiprazole for 30 min at 37 °C. The metabolite DM-3411 was detected using ultraperformance liquid chromatography-tandem mass spectrometry. The activity of the wild-type CYP3A4 and 26 of its variants was analyzed. Then, the mechanism underlying the changes in enzyme function was observed using molecular dynamics simulations and molecular docking. Compared with CYP3A4.1, the enzymatic activities of CYP3A4.19, -.24, and -.28 were not significantly different (from 91.82% to 96.25%), but CYP3A4.14 and CYP3A4.15 exhibited higher enzyme activity (from 117.9 to 127.5%). The remaining 21 isoforms, including CYP3A4.2, -.3, -.4, -.5, -.7, -.8, -.9, -.10, -.11, -.12, -.13, -.16, -.17, -.18, -.20, -.23, -.29, -.31, -.32, -.33 and -.34, displayed lower enzymatic activities (from 2.90% to 75.72%). The results obtained from computer modeling indicated that weak binding affinity impaired the function of CYP3A4.32. Mutations that occur around the active site might lead to a loss of enzymatic activity, while the variants located far away from the active site perhaps had little effect on function of CYP3A4. These comprehensive data provide a reference and prediction for treatment strategies and risk assessments of brexpiprazole.


Subject(s)
Antipsychotic Agents/metabolism , Cytochrome P-450 CYP3A/metabolism , Dopamine Agonists/metabolism , Quinolones/metabolism , Serotonin Agents/metabolism , Thiophenes/metabolism , Cytochrome P-450 CYP3A/genetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Recombinant Proteins/metabolism
4.
Pharmacology ; 105(1-2): 79-89, 2020.
Article in English | MEDLINE | ID: mdl-31587004

ABSTRACT

WZ35 is a monocarbonyl analog of curcumin, which had been proved advantage over curcumin in chemical stability and antitumor activity. However, its pharmacokinetic profile has not been determined. In the present study, an ultraperformance liquid chromatography-tandem mass spectrometry assay was developed to detect concentration of WZ35 in rat plasma. Subsequently, pharmacokinetic study showed that the oral bioavailability of WZ35 is 10.56%. Cytochrome P450 (CYP450) plays a major role in metabolizing exogenous substance. The concentration of WZ35 was sharply decreased while incubating with microsome. It's indicated that WZ35 is a substrate of CYP450s. Molecular docking assay showed that WZ35 can combine with CYP2B6 and CYP2C9 to form much more stable complex. The lowest docking energy was generated in complex with CYP2E1. The inhibition of CYP450s by WZ35 was also evaluated. Pan inhibitions of WZ35 on rat CYP3A2, CYP2B1, CYP2C11, CYP2D1, and -CYP2E1 were observed by detecting probe substrates (midazolam, bupropion, tolbutamide, dextromethorphan, chlorzoxazone) and metabolites accordingly. On an average, 80% activities of enzymes were blocked. Mechanistically, the inhibitions of WZ35 on CYP3A2, CYP2B1, CYP2E1 were in a time-dependent manner according to the results of IC50 shift assay. The collective data demonstrated that the oral bioavailability of monocarbonyl analog of curcumin has significantly improved compared to curcumin. It's both the substrate and inhibitor of CYP450s through in a time-dependent mechanism.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Curcumin/analogs & derivatives , Curcumin/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Animals , Antineoplastic Agents/blood , Biological Availability , Cytochrome P-450 Enzyme Inhibitors/blood , Cytochrome P-450 Enzyme System/metabolism , Male , Molecular Docking Simulation , Rats, Sprague-Dawley
5.
Eur J Med Chem ; 233: 114249, 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35259690

ABSTRACT

Pan-HER inhibitors exhibit extensive biological activity and offer unique advantages and usually bind to targets in an irreversible manner. Owing to the off-target toxicity of irreversible inhibitors, reversible pan-HER inhibitors are desirable. Herein, we describe the process of N-(ring structure fused phenyl)quinazoline-4-amine-based design, synthesis, and biological evaluation of pan-HER inhibitors in vitro and in vivo. Compound C5, with the molecular skeleton of N-(3-bromo-1H-indol-5-yl)-quinazolin-4-amine, displayed irreversible binding just like other effective pan-HER inhibitors. To our surprise, compound C6, which possessed the same skeleton, was found to be a high-strength reversible pan-HER inhibitor. This compound was capable of inhibiting HER1s (such as EGFR T790M/L858R and WT), HER2, and HER4 and can be considered as a breakthrough in the development of pan-HER inhibitors. Altogether, N-(3-bromo-1H-indol-5-yl)-quinazolin-4-amine can serve as an effective molecular skeleton for developing both reversible and irreversible pan-HER inhibitors in the following discovery of antitumor drugs.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Quinazolines/chemistry , Quinazolines/pharmacology , Cell Line, Tumor , ErbB Receptors/genetics , Humans , Molecular Structure , Mutation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
6.
Chem Biol Interact ; 345: 109559, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34153224

ABSTRACT

AIM: We aimed (i) to study the effects of genetic polymorphism of cytochrome P450 3A4 (CYP3A4) and drug interactions on acalabrutinib (ACA) metabolism and (ii) to investigate the mechanisms underlying the effects of CYP3A4 variants on the differential kinetic profiles of ACA and ibrutinib. METHOD: Recombinant human CYP3A4 and variants were expressed using a Bac-to-Bac baculovirus expression system. The cell microsome was prepared and subjected to kinetic study. The analyte concentrations were determined by UPLC-MS/MS. A molecular docking assay was employed to investigate the mechanisms leading to differences in kinetic profiles. RESULTS: The kinetic parameters of ACA, catalyzed by CYP3A4 and 28 of its variants, were determined, including Vmax, Km, and Ksi. CYP3A4.6-8, 12, 13, 17, 18, 20, and 30 lost their catalytic function. No significant differences were found for CYP3A4.4, 5, 10, 15, 31, and 34 compared with CYP3A4.1 with respect to intrinsic clearance (Vmax/Km, Clint). However, the Clint values of CYP3A4.9, 14, 16, 19, 23, 24, 28, 32 were obviously decreased, ranging from 0.02 to 0.05 µL/min/pmol. On the contrary, the catalytic activities of CYP3A4.2, 3, 11, 29, and 33 were increased dramatically. The Clint value of CYP3A4.11 was 5.95 times as high as that of CYP3A4.1. Subsequently, CYP3A4.1, 3, 11, 23, and 28 were chosen to study the kinetic changes in combination with ketoconazole. Interestingly, we found the inhibitory potency of ketoconazole varied in different variants. In addition, the kinetic parameters of ibrutinib and ACA were accordingly compared in different CYP3A4 variants. Significant differences in relative clearance were observed among variants, which would probably influence the distance between the redox site and the heme iron atom. CONCLUSION: Genetic polymorphism of CYP3A4 extensively changes its ACA-metabolizing enzymatic activity. In combination with a CYP inhibitor, its inhibitory potency also varied among different variants. Even the same variants exhibited different capabilities catalyzing ACA. Its enzymatic capabilities are probably determined by the distance between the substrate and the heme iron atom, which could be impacted by mutation.


Subject(s)
Benzamides/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Genetic Variation , Pyrazines/metabolism , Biocatalysis , Cytochrome P-450 CYP3A/chemistry , Heme/metabolism , Humans , Molecular Docking Simulation , Oxidation-Reduction , Protein Conformation
7.
Front Pharmacol ; 12: 775117, 2021.
Article in English | MEDLINE | ID: mdl-34912226

ABSTRACT

Myeloid differentiation factor 88 (MyD88) is a hub protein in the Toll-like receptor signaling pathway, which acts as a master switch for numerous inflammatory diseases, including acute lung injury (ALI). Although this protein is considered as a crucial therapeutic target, there are currently no clinically approved MyD88-targeting drugs. Based on previous literature, here we report the discovery via computer-aided drug design (CADD) of a small molecule, M20, which functions as a novel MyD88 inhibitor to efficiently relieve lipopolysaccharide-induced inflammation both in vitro and in vivo. Computational chemistry, surface plasmon resonance detection (SPR) and biological experiments demonstrated that M20 forms an important interaction with the MyD88-Toll/interleukin-1 receptor domain and thereby inhibits the protein dimerization. Taken together, this study found a MyD88 inhibitor, M20, with a novel skeleton, which provides a crucial understanding in the development and modification of MyD88 inhibitors. Meanwhile, the favorable bioactivity of the hit compound is also conducive to the treatment of acute lung injury or other more inflammatory diseases.

8.
Arthritis Res Ther ; 23(1): 266, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702315

ABSTRACT

BACKGROUND: Connective tissue growth factor (CTGF)-induced angiogenesis is a crucial factor in rheumatoid arthritis (RA), but CTGF-interacting protein and related molecular mechanism of their interaction have not been fully elucidated. METHODS: CTGF-interacting proteins were identified through the LC-MS/MS analysis of the Co-IP products from fibroblast-like synoviocyte (FLS) lysates, and the interaction between CTGF and annexin A2 (ANXA2) was further confirmed through Co-IP and BiFC assay. The binding domain, mutant, mechanism, and angiogenesis function were assessed by homology modeling, molecular docking, MTT, cell scratch, tube formation, and chick chorioallantoic membrane (CAM) assays. Additionally, severe combined immunodeficiency (SCID) mouse co-implantation model was constructed to confirm the effect of ANXA2/CTGF-TSP1 in the process of RA in vivo. RESULTS: ANXA2 was identified and verified as an interaction partner of CTGF for the first time by Co-IP and LC-MS/MS analysis. Co-localization of CTGF and ANXA2 was observed in RA-FLS, and direct interaction of the TSP-1 domain of CTGF and ANXA2 was determined in HEK293T cells. The spatial conformation and stable combination of the ANXA2/CTGF-TSP1 complex were assessed by homology modeling in the biomimetic environment. The function of the ANXA2/CTGF-TSP1 complex was proved on promoting FLS proliferation, migration, and angiogenesis in vitro and deteriorating FLS invasion and joint damage in SCID mice. CONCLUSIONS: TSP-1 is the essential domain in CTGF/ANXA2 interaction and contributes to FLS migration and pannus formation, inducing the process of RA.


Subject(s)
Annexin A2 , Arthritis, Rheumatoid , Animals , Annexin A2/genetics , Chromatography, Liquid , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , HEK293 Cells , Humans , Mice , Molecular Docking Simulation , Pannus , Tandem Mass Spectrometry
9.
Eur J Med Chem ; 214: 113219, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33618175

ABSTRACT

Fibroblast growth factor receptor 4 (FGFR4) is a member of the fibroblast growth factor receptor family, which is closely related to the occurrence and development of hepatocellular carcinoma (HCC). In this article, a series of indazole derivatives were designed and synthesized by using computer-aided drug design (CADD) and structure-based design strategies, and then they were evaluated for their inhibition of FGFR4 kinase and antitumor activity. F-30 was subtly selective for FGFR4 compared to FGFR1; it affected cell growth and migration by inhibiting FGFR4 pathways in HCC cell lines in a dose-dependent manner.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Design , Indazoles/pharmacology , Liver Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indazoles/chemical synthesis , Indazoles/chemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Receptor, Fibroblast Growth Factor, Type 4 , Structure-Activity Relationship
10.
Front Cell Dev Biol ; 8: 580517, 2020.
Article in English | MEDLINE | ID: mdl-33072762

ABSTRACT

Colon cancer is one of the leading causes of cancer-related death in the world. The development of new drugs and therapeutic strategies for patients with colon cancer are urgently needed. Isodeoxyelephantopin (ESI), a sesquiterpene lactone isolated from the medicinal plant Elephantopus scaber L., has been reported to exert antitumor effects on several cancer cells. However, the molecular mechanisms underlying the action of ESI is still elusive. In the present study, we found that ESI potently suppressed cell proliferation in human colon cancer cells. Furthermore, our results showed that ESI treatment markedly increased cellular reactive oxygen species (ROS) levels by inhibiting thioredoxin reductase 1 (TrxR1) activity, which leads to activation of the JNK signaling pathway and eventually cell death in HCT116 and RKO cells. Importantly, we found that ESI markedly enhanced cisplatin-induced cytotoxicity in HCT116 and RKO cells. Combination of ESI and cisplatin significantly increased the production of ROS, resulting in activation of the JNK signaling pathway in HCT116 and RKO cells. In vivo, we found that ESI combined with cisplatin significantly suppressed tumor growth in HCT116 xenograft models. Together, our study provide a preclinical proof-of-concept for ESI as a potential strategy for colon cancer treatment.

11.
J Pharm Pharmacol ; 72(10): 1405-1411, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32608074

ABSTRACT

OBJECTIVE: In this study, we aimed to investigate the potential interaction of apatinib and buspirone and underlying mechanism. METHODS: UPLC-MS/MS assay was applied to determine the concentrations of buspirone and its main metabolites (1-PP and 6-OH buspirone) after incubated with liver microsomes. Moreover, the connection of in vitro and in vivo was further determined. Sprague Dawley rats were randomly divided into two groups: group A (20 mg/kg buspirone) and group B (buspirone vs 40 mg/kg apatinib). Tail vein blood was collected and subjected to the UPLC-MS/MS detection. KEY FINDINGS: Apatinib inhibited the generations of 1-PP and 6-OH buspirone dose-dependently with IC50 of 1.76 and 2.23 µm in RLMs, and 1.51 and 1.48 µm in HLMs, respectively. There was a mixed mechanism underlying such an inhibition effect. In rat, AUC(0- t ) , AUC(0-∞) , Tmax and Cmax of buspirone and 6-OH buspirone increased significantly while co-administering with apatinib, but Vz/F and CLz/F decreased obviously while comparing group A with group B . CONCLUSIONS: Apatinib suppresses the CYP450 based metabolism of buspirone in a mixed mechanism and boosted the blood exposure of prototype drug and 6-OH buspirone dramatically. Therefore, extra caution should be taken when combining apatinib with buspirone in clinic.


Subject(s)
Buspirone/pharmacokinetics , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/pharmacokinetics , Animals , Buspirone/administration & dosage , Dose-Response Relationship, Drug , Drug Interactions/physiology , Male , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Rats , Rats, Sprague-Dawley
12.
Int J Biol Macromol ; 135: 294-302, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31128178

ABSTRACT

Novel efficacious treatment of Rhabdomyosarcoma (RMS) with less toxicity has yet to emerge. Genomic analysis of RMS has reported that the receptor tyrosine kinase FGFR4 is highly expressed and frequently mutated in the tumor tissue. The V550E/L and N535D/K mutations of FGFR4 in RMS can lead to strong drug resistance to almost all of the type-I inhibitors. Previous report has demonstrated the type-II inhibitor ponatinib is the most potentially effective agent for RMS but still hard to starboard the V550E/L mutants. In this case, an ensemble of molecular modeling strategies was employed to theoretically uncover the resistance mechanisms. The binding free energy calculation results predicted by various strategies show that the V550E/L rather than N535D/K mutations indeed weaken the binding affinity of ponatinib, which are in good agreement with the experimental observations. Subsequently, the energy decomposition analysis mapped a knock-on effect on the diverse energy components of some key residues. Moreover, it is of great importance to report that there is an effective channel for type-II inhibitors sliding along the A-loop to prevent FGFR4 from phosphorylation and activation. Our results provide new insight into drug binding process and guide the development of effective inhibitors to surmount drug resistance in RMS.


Subject(s)
Antineoplastic Agents/chemistry , Drug Resistance, Neoplasm/genetics , Imidazoles/chemistry , Models, Molecular , Mutation , Protein Kinase Inhibitors/chemistry , Pyridazines/chemistry , Receptor, Fibroblast Growth Factor, Type 4/chemistry , Receptor, Fibroblast Growth Factor, Type 4/genetics , Amino Acid Substitution , Antineoplastic Agents/pharmacology , Binding Sites , Computers, Molecular , Humans , Imidazoles/pharmacology , Protein Binding , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Rhabdomyosarcoma/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL