Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochem Pharmacol ; 224: 116230, 2024 06.
Article in English | MEDLINE | ID: mdl-38643905

ABSTRACT

One of the effective therapeutic strategies to treat rheumatoid arthritis (RA)-related bone resorption is to target excessive activation of osteoclasts. We discovered that 6-O-angeloylplenolin (6-OAP), a pseudoguaianolide from Euphorbia thymifolia Linn widely used for the treatment of RA in traditional Chinese medicine, could inhibit RANKL-induced osteoclastogenesis and bone resorption in both RAW264.7 cells and BMMs from 1 µM and protect a collagen-induced arthritis (CIA) mouse model from bone destruction in vivo. The severity of arthritis and bone erosion observed in paw joints and the femurs of the CIA model were attenuated by 6-OAP administered at both dosages (1 or 5 mg/kg, i.g.). BMD, Tb.N and BV/TV were also improved by 6-OAP treatment. Histological analysis and TRAP staining of femurs further confirmed the protective effects of 6-OAP on bone erosion, which is mainly due to reduced osteoclasts. Molecular docking indicated that c-Src might be a target of 6-OAP and phosphorylation of c-Src was suppressed by 6-OAP treatment. CETSA and SPR assay further confirmed the potential interaction between 6-OAP and c-Src. Three signaling molecules downstream of c-Src that are vital to the differentiation and function of osteoclasts, NF-κB, c-Fos and NFATc1, were also suppressed by 6-OAP in vitro. In summary, the results demonstrated that the function of c-Src was disrupted by 6-OAP, which led to the suppression of downstream signaling vital to osteoclast differentiation and function. In conclusion, 6-OAP has the potential to be further developed for the treatment of RA-related bone erosion.


Subject(s)
Arthritis, Experimental , Bone Resorption , NF-kappa B , NFATC Transcription Factors , Osteoclasts , Osteogenesis , Animals , Mice , NFATC Transcription Factors/metabolism , RAW 264.7 Cells , Bone Resorption/drug therapy , Bone Resorption/metabolism , Bone Resorption/prevention & control , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Arthritis, Experimental/chemically induced , Osteogenesis/drug effects , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Male , Signal Transduction/drug effects , CSK Tyrosine-Protein Kinase/metabolism , Molecular Docking Simulation , src-Family Kinases/metabolism , src-Family Kinases/antagonists & inhibitors
2.
BMC Cancer ; 22(1): 256, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35272617

ABSTRACT

BACKGROUND: Over half of colorectal cancers (CRCs) are hard-wired to RAS/RAF/MEK/ERK pathway oncogenic signaling. However, the promise of targeted therapeutic inhibitors, has been tempered by disappointing clinical activity, likely due to complex resistance mechanisms that are not well understood. This study aims to investigate MEK inhibitor-associated resistance signaling and identify subpopulation(s) of CRC patients who may be sensitive to biomarker-driven drug combination(s). METHODS: We classified 2250 primary and metastatic human CRC tumors by consensus molecular subtypes (CMS). For each tumor, we generated multiple gene expression signature scores measuring MEK pathway activation, MEKi "bypass" resistance, SRC activation, dasatinib sensitivity, EMT, PC1, Hu-Lgr5-ISC, Hu-EphB2-ISC, Hu-Late TA, Hu-Proliferation, and WNT activity. We carried out correlation, survival and other bioinformatic analyses. Validation analyses were performed in two independent publicly available CRC tumor datasets (n = 585 and n = 677) and a CRC cell line dataset (n = 154). RESULTS: Here we report a central role of SRC in mediating "bypass"-resistance to MEK inhibition (MEKi), primarily in cancer stem cells (CSCs). Our integrated and comprehensive gene expression signature analyses in 2250 CRC tumors reveal that MEKi-resistance is strikingly-correlated with SRC activation (Spearman P < 10-320), which is similarly associated with EMT (epithelial to mesenchymal transition), regional metastasis and disease recurrence with poor prognosis. Deeper analysis shows that both MEKi-resistance and SRC activation are preferentially associated with a mesenchymal CSC phenotype. This association is validated in additional independent CRC tumor and cell lines datasets. The CMS classification analysis demonstrates the strikingly-distinct associations of CMS1-4 subtypes with the MEKi-resistance and SRC activation. Importantly, MEKi + SRCi sensitivities are predicted to occur predominantly in the KRAS mutant, mesenchymal CSC-like CMS4 CRCs. CONCLUSIONS: Large human tumor gene expression datasets representing CRC heterogeneity can provide deep biological insights heretofore not possible with cell line models, suggesting novel repurposed drug combinations. We identified SRC as a common targetable node--an Achilles' heel--in MEKi-targeted therapy-associated resistance in mesenchymal stem-like CRCs, which may help development of a biomarker-driven drug combination (MEKi + SRCi) to treat problematic subpopulations of CRC.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , src-Family Kinases/antagonists & inhibitors , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/drug effects , Humans , MAP Kinase Signaling System/drug effects , Proto-Oncogene Proteins p21(ras)/genetics , Transcriptome/drug effects
3.
Phytomedicine ; 95: 153705, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34538671

ABSTRACT

BACKGROUND: Liver cancer is one of the leading causes of cancer-related death worldwide. Dihydrotanshinone I (DHI) was shown to inhibit the growth of several types of cancer. However, research related to hepatoma treatment using DHI is limited. PURPOSE: Here, we explored the inhibitory effect of DHI on the growth of hepatoma cells, and investigated the underlying molecular mechanisms. METHODS: The proliferation of Hep3B, SMCC-7721 and SK-Hep1 hepatoma cells was evaluated using the MTS and Edu staining assay. Hepatoma cell death was analyzed with a LIVE/DEAD Cell Imaging Kit. The relative expression and phosphorylation of proto-oncogene tyrosine-protein kinase Src (Src) and signal transducer and activator of transcription-3 (STAT3) proteins in hepatoma cells, as well as the expression of other protein components, were measured by western blotting. The structural interaction of DHI with Src proteins was evaluated by molecular docking, molecular dynamics simulation, surface plasmon resonance imaging and Src kinase inhibition assay. Src overexpression was achieved by infection with an adenovirus vector encoding human Src. Subsequently, the effects of DHI on tumor growth inhibition were further validated using mouse xenograft models of hepatoma. RESULTS: In vitro studies showed that treatment with DHI inhibited the proliferation and promoted cell death of Hep3B, SMCC-7721 and SK-Hep1 hepatoma cells. We further identified and verified Src as a direct target of DHI by using molecular stimulation, surface plasmon resonance image and Src kinase inhibition assay. Treatment with DHI reduced the in vitro phosphorylation levels of Src and STAT3, a transcription factor regulated by Src. In the xenograft mouse models, DHI dose-dependently suppressed tumor growth and Src and STAT3 phosphorylation. Moreover, Src overexpression partly abrogated the inhibitory effects of DHI on the proliferation and cell death in hepatoma cells. CONCLUSION: Our results suggest that DHI inhibits the growth of hepatoma cells by direct inhibition of Src.


Subject(s)
Carcinoma, Hepatocellular , Furans/pharmacology , Phenanthrenes , Quinones/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation , Mice , Molecular Docking Simulation , Phenanthrenes/pharmacology , Phosphorylation , STAT3 Transcription Factor/metabolism , src-Family Kinases/metabolism
4.
Pharm Biol ; 59(1): 799-810, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34190667

ABSTRACT

CONTEXT: Among the plants in the genus Barringtonia (Lecythidaceae) used as traditional medicines to treat arthralgia, chest pain, and haemorrhoids in Indonesia, Barringtonia racemosa L. and Barringtonia acutangula (L.) Gaertn. have demonstrated anti-inflammatory activity in systemic inflammatory models. OBJECTIVE: The anti-inflammatory activity of Barringtonia angusta Kurz has not been investigated. We prepared a methanol extract of the leaves and stems of B. angusta (Ba-ME) and systemically evaluated its anti-inflammatory effects in vitro and in vivo. MATERIALS AND METHODS: RAW264.7 cells stimulated with LPS or Pam3CSK4 for 24 h were treated with Ba-ME (12.5, 25, 50, 100, and 150 µg/mL), and NO production and mRNA levels of inflammatory genes were evaluated. Luciferase reporter gene assay, western blot analysis, overexpression experiments, and cellular thermal shift assay were conducted to explore the mechanism of Ba-ME. In addition, the anti-gastritis activity of Ba-ME (50 and 100 mg/kg, administered twice per day for two days) was evaluated using an HCl/EtOH-induced gastritis mouse model. RESULTS: Ba-ME dose-dependently suppressed NO production [IC50 = 123.33 µg/mL (LPS) and 46.89 µg/mL (Pam3CSK4)] without affecting cell viability. Transcriptional expression of iNOS, IL-1ß, COX-2, IL-6, and TNF-α and phosphorylation of Src, IκBα, p50/105, and p65 were inhibited by Ba-ME. The extract specifically targeted the Src protein by binding to its SH2 domain. Moreover, Ba-ME significantly ameliorated inflammatory lesions in the HCl/EtOH-induced gastritis model. DISCUSSION AND CONCLUSIONS: The anti-inflammatory activity of Ba-ME is mediated by targeting of the Src/NF-κB signalling pathway, and B. angusta has potential as an anti-inflammatory drug.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Barringtonia , Drug Delivery Systems/methods , Gastritis/drug therapy , Plant Extracts/administration & dosage , src-Family Kinases/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/metabolism , Dose-Response Relationship, Drug , Gastritis/chemically induced , Gastritis/metabolism , HEK293 Cells , Humans , Male , Methanol/administration & dosage , Methanol/metabolism , Mice , Mice, Inbred ICR , NF-kappa B , Plant Extracts/isolation & purification , Plant Extracts/metabolism , Plant Leaves , Plant Stems , RAW 264.7 Cells , Signal Transduction/drug effects , Signal Transduction/physiology , src-Family Kinases/metabolism
5.
Pharm Biol ; 59(1): 74-86, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33439064

ABSTRACT

CONTEXT: Sauropus brevipes Müll. Arg. (Phyllanthaceae) has been used as an effective ingredient in a decoction for the treatment of diarrhoea. However, there was no report on its modulatory role in inflammation. OBJECTIVE: This study investigates anti-inflammatory effect of S. brevipes in various inflammation models. MATERIALS AND METHODS: The aerial part of S. brevipes was extracted with 95% ethanol to produce Sb-EE. RAW264.7 cells pre-treated with Sb-EE were stimulated by lipopolysaccharide (LPS), and Griess assay and PCR were performed. High-performance liquid chromatography (HPLC) analysis, luciferase assay, Western blotting and kinase assay were employed. C57BL/6 mice (10 mice/group) were orally administered with Sb-EE (200 mg/kg) once a day for five days, and peritonitis was induced by an intraperitoneal injection of LPS (10 mg/kg). ICR mice (four mice/group) were orally administered with Sb-EE (20 or 200 mg/kg) or ranitidine (positive control) twice a day for two days, and EtOH/HCl was orally injected to induce gastritis. RESULTS: Sb-EE suppressed nitric oxide (NO) release (IC50=34 µg/mL) without cytotoxicity and contained flavonoids (quercetin, luteolin and kaempferol). Sb-EE (200 µg/mL) reduced the mRNA expression of inducible NO synthase (iNOS). Sb-EE blocked the activities of Syk and Src, while inhibiting interleukin-1 receptor associated kinases (IRAK1) by 68%. Similarly, orally administered Sb-EE (200 mg/kg) suppressed NO production by 78% and phosphorylation of Src and Syk in peritonitis mice. Sb-EE also decreased inflammatory lesions in gastritis mice. DISCUSSION AND CONCLUSIONS: This study demonstrates the inhibitory effect of Sb-EE on the inflammatory response, suggesting that Sb-EE can be developed as a potential anti-inflammatory agent.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drug Delivery Systems/methods , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Plant Extracts/therapeutic use , Syk Kinase/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/therapeutic use , Ethanol/pharmacology , Ethanol/therapeutic use , Gastritis/drug therapy , Gastritis/metabolism , HEK293 Cells , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Peritonitis/drug therapy , Peritonitis/metabolism , Plant Components, Aerial , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , RAW 264.7 Cells , Syk Kinase/metabolism , src-Family Kinases/metabolism
6.
Phytomedicine ; 80: 153391, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33113502

ABSTRACT

BACKGROUND: Pseudo-allergic reactions are potentially fatal hypersensitivity responses caused by mast cell activation. α-linolenic acid (ALA) is known for its anti-allergic properties. However, its potential anti-pseudo-allergic effects were not much investigated. PURPOSE: To investigate the inhibitory effects of ALA on IgE-independent allergy in vitro, and in vivo, as well as the mechanism underlying its effects. METHODS/STUDY DESIGNS: The anti-anaphylactoid activity of ALA was evaluated in passive cutaneous anaphylaxis reaction (PCA) and systemic anaphylaxis models. Calcium imaging was used to assess intracellular Ca2+ mobilization. The release of cytokines and chemokines was measured using enzyme immunoassay kits. Western blot analysis was conducted to investigate the molecules of Lyn-PLCγ-IP3R-Ca2+ and Lyn-p38/NF-κB signaling pathway. RESULTS: ALA (0, 1.0, 2.0, and 4.0 mg/kg) dose-dependently reduced serum histamine, chemokine release, vasodilation, eosinophil infiltration, and the percentage of degranulated mast cells in C57BL/6 mice. In addition, ALA (0, 50, 100, and 200 µM) reduced Compound 48/80 (C48/80) (30 µg/ml)-or Substance P (SP) (4 µg/ml)-induced calcium influx, mast cell degranulation and cytokines and chemokine release in Laboratory of Allergic Disease 2 (LAD2) cells via Lyn-PLCγ-IP3R-Ca2+ and Lyn-p38/NF-κB signaling pathway. Moreover, ALA (0, 50, 100, and 200 µM) inhibited C48/80 (30 µg/ml)- and SP (4 µg/ml)-induced calcium influx in Mas-related G-protein coupled receptor member X2 (MrgX2)-HEK293 cells and in vitro kinase assays confirmed that ALA inhibited the activity of Lyn kinase. In response to 200 µM of ALA, the activity of Lyn kinase by (7.296 ± 0.03751) × 10-5 units/µl and decreased compared with C48/80 (30 µg/ml) by (8.572 ± 0.1365) ×10-5 units/µl. CONCLUSION: Our results demonstrate that ALA might be a potential Lyn kinase inhibitor, which could be used to treat pseudo-allergic reaction-related diseases such as urticaria.


Subject(s)
Anaphylaxis/drug therapy , Anti-Allergic Agents/pharmacology , Passive Cutaneous Anaphylaxis/drug effects , alpha-Linolenic Acid/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Cell Degranulation/drug effects , Chemokines/metabolism , Dose-Response Relationship, Drug , Humans , Immunoglobulin E/immunology , Male , Mast Cells/drug effects , Mast Cells/immunology , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , p-Methoxy-N-methylphenethylamine/toxicity , src-Family Kinases/chemistry , src-Family Kinases/immunology , src-Family Kinases/metabolism
7.
J Cell Physiol ; 236(2): 1148-1157, 2021 02.
Article in English | MEDLINE | ID: mdl-32686156

ABSTRACT

Saracatinib is an oral Src-kinase inhibitor and has been studied in preclinical models and clinical trials of cancer therapy. GMI, a fungal immunomodulatory protein from Ganoderma microsporum, possesses antitumor capacity. The aim of this study is to evaluate the cytotoxic effect of combination treatment with saracatinib and GMI on parental and pemetrexed-resistant lung cancer cells. Cotreatment with saracatinib and GMI induced synergistic and additive cytotoxic effect in A549 and A400 cells by annexin V/propidium iodide assay and combination index. Using western blot assay, saracatinib, and GMI combined treatment synergistically induced caspase-7 activation in A549 cells. Different from A549 cells, saracatinib and GMI cotreatment markedly increased LC3B-II in A400 cells. ATG5 silencing abolished the caspase-7 activation and reduced cell death in A549 cells after cotreatment. This is the first study to provide a novel strategy of treating lung cancer with or without drug resistance via combination treatment with GMI and saracatinib.


Subject(s)
Autophagy-Related Protein 5/genetics , Benzodioxoles/pharmacology , Caspase 7/genetics , Enzyme Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Quinazolines/pharmacology , src-Family Kinases/genetics , A549 Cells , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5/antagonists & inhibitors , Cell Proliferation/drug effects , Fungal Proteins/chemistry , Fungal Proteins/pharmacology , Ganoderma/chemistry , Humans , Immunologic Factors/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Synthetic Lethal Mutations/drug effects , Xenograft Model Antitumor Assays , src-Family Kinases/antagonists & inhibitors
8.
Acta Pharmacol Sin ; 42(3): 370-381, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33303991

ABSTRACT

Stroke is an acute cerebrovascular disease caused by ruptured or blocked blood vessels. For the prevention of ischemic stroke, the coagulation state of blood and cerebrovascular protection should be considered. Our previous study has shown that salvianolic acid A (SAA), which is a water-soluble component from the root of Salvia Miltiorrhiza Bge, prevents thrombosis with a mild inhibitory effect on platelet aggregation. In this study we investigated the preventive effects of SAA on cerebrovascular endothelial injury caused by ischemia in vivo and oxygen-glucose deprivation (OGD) in vitro, and explored the underlying mechanisms. An autologous thrombus stroke model was established in SD rats by electrocoagulation. SAA (10 mg/kg) was orally administered twice a day for 5 days before the operation. The rats were sacrificed at 24 h after the operation. We showed that pretreatment with SAA significantly improved the neurological deficits, intracerebral hemorrhage, BBB disruption, and vascular endothelial dysfunction as compared with model group. In human brain microvascular endothelial cells (HBMECs), pretreatment with SAA (10 µM) significantly inhibited OGD-induced cell viability reduction and degradation of tight junction proteins (ZO-1, occludin, claudin-5). Furthermore, we found that SAA inhibited the upregulation of Src signaling pathway in vivo and vitro and reversed the increased expression of matrix metalloproteinases (MMPs) after ischemic stroke. In conclusion, our results suggest that SAA protects cerebrovascular endothelial cells against ischemia and OGD injury via suppressing Src signaling pathway. These findings show that pretreatment with SAA is a potential therapeutic strategy for the prevention of ischemic stroke.


Subject(s)
Caffeic Acids/therapeutic use , Endothelium, Vascular/drug effects , Ischemic Stroke/prevention & control , Lactates/therapeutic use , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , Animals , Blood-Brain Barrier/drug effects , Brain/blood supply , Brain/drug effects , Cerebral Hemorrhage/prevention & control , Enzyme Activation/drug effects , Humans , Male , Rats, Sprague-Dawley , Tight Junctions/drug effects , src-Family Kinases/antagonists & inhibitors
9.
Biomed Res Int ; 2020: 5891016, 2020.
Article in English | MEDLINE | ID: mdl-33145355

ABSTRACT

BACKGROUND: Kaempferol is a natural polyphenol in lots of Chinese herbs, which has shown promising treatment for gastric cancer (GC). However, the molecular mechanisms of its action have not been systematically revealed yet. In this work, a network pharmacology approach was used to elucidate the potential mechanisms of kaempferol in the treatment of GC. METHODS: The kaempferol was input into the PharmMapper and SwissTargetPrediction database to get its targets, and the targets of GC were obtained by retrieving the Online Mendelian Inheritance in Man (OMIM) database, MalaCards database, Therapeutic Target Database (TTD), and Coolgen database. The molecular docking was performed to assess the interactions between kaempferol and these targets. Next, the overlap targets of kaempferol and GC were identified for GO and KEGG enrichment analyses. Afterward, a protein-protein interaction (PPI) network was constructed to get the hub targets, and the expression and overall survival analysis of the hub target were investigated. Finally, the overall survival (OS) analysis of hub targets was performed using the Kaplan-Meier Plotter online tool. RESULTS: A total of 990 genes related to GC and 10 overlapping genes were determined through matching the 24 potential targets of kaempferol with disease-associated genes. The result of molecular docking indicated that kaempferol can bind with these hub targets with good binding scores. These targets were further mapped to 140 GO biological process terms and 11 remarkable pathways. In the PPI network analysis, 3 key targets were identified, including ESR1, EGFR, and SRC. The mRNA and protein expression levels of EGFR and SRC were obviously higher in GC tissues. High expression of these targets was related to poor OS in GC patients. CONCLUSIONS: This study provided a novel approach to reveal the therapeutic mechanisms of kaempferol on GC, which will ease the future clinical application of kaempferol in the treatment of GC.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Kaempferols/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Stomach Neoplasms/drug therapy , src-Family Kinases/antagonists & inhibitors , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aged , Binding Sites , Drugs, Chinese Herbal , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Molecular Docking Simulation , Molecular Targeted Therapy/methods , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pharmacogenetics , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Interaction Maps , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Analysis , src-Family Kinases/chemistry , src-Family Kinases/genetics , src-Family Kinases/metabolism
10.
Molecules ; 25(18)2020 Sep 08.
Article in English | MEDLINE | ID: mdl-32911607

ABSTRACT

Src plays a crucial role in many signaling pathways and contributes to a variety of cancers. Therefore, Src has long been considered an attractive drug target in oncology. However, the development of Src inhibitors with selectivity and novelty has been challenging. In the present study, pharmacophore-based virtual screening and molecular docking were carried out to identify potential Src inhibitors. A total of 891 molecules were obtained after pharmacophore-based virtual screening, and 10 molecules with high docking scores and strong interactions were selected as potential active molecules for further study. Absorption, distribution, metabolism, elimination and toxicity (ADMET) property evaluation was used to ascertain the drug-like properties of the obtained molecules. The proposed inhibitor-protein complexes were further subjected to molecular dynamics (MD) simulations involving root-mean-square deviation and root-mean-square fluctuation to explore the binding mode stability inside active pockets. Finally, two molecules (ZINC3214460 and ZINC1380384) were obtained as potential lead compounds against Src kinase. All these analyses provide a reference for the further development of novel Src inhibitors.


Subject(s)
Drug Discovery , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , src-Family Kinases/chemistry , Binding Sites , Databases, Pharmaceutical , Drug Discovery/methods , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Conformation , Molecular Structure , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Quantitative Structure-Activity Relationship , Reproducibility of Results , src-Family Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL