Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 550
Filter
1.
Dis Model Mech ; 17(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39051113

ABSTRACT

Hepatocellular carcinoma (HCC) is a disease of high unmet medical need that has become a global health problem. The development of targeted therapies for HCC has been hindered by the incomplete understanding of HCC pathogenesis and the limited number of relevant preclinical animal models. We recently unveiled a previously uncharacterized YES kinase (encoded by YES1)-dependent oncogenic signaling pathway in HCC. To model this subset of HCC, we established a series of syngeneic cell lines from liver tumors of transgenic mice expressing activated human YES. The resulting cell lines (referred to as HepYF) were enriched for expression of stem cell and progenitor markers, proliferated rapidly, and were characterized by high SRC family kinase (SFK) activity and activated mitogenic signaling pathways. Transcriptomic analysis indicated that HepYF cells are representative of the most aggressive proliferation class G3 subgroup of HCC. HepYF cells formed rapidly growing metastatic tumors upon orthotopic implantation into syngeneic hosts. Treatment with sorafenib or the SFK inhibitor dasatinib markedly inhibited the growth of HepYF tumors. The new HepYF HCC cell lines provide relevant preclinical models to study the pathogenesis of HCC and test novel small-molecule inhibitor and immunotherapy approaches.


Subject(s)
Carcinoma, Hepatocellular , Cell Proliferation , Disease Models, Animal , Liver Neoplasms , Neoplasm Metastasis , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Cell Proliferation/drug effects , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Humans , Cell Line, Tumor , Sorafenib/pharmacology , Sorafenib/therapeutic use , Dasatinib/pharmacology , Dasatinib/therapeutic use , Mice, Transgenic , Mice , src-Family Kinases/metabolism , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Signal Transduction/drug effects , Niacinamide/analogs & derivatives , Niacinamide/pharmacology
2.
Anticancer Res ; 44(8): 3287-3294, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060052

ABSTRACT

BACKGROUND/AIM: Transforming growth factor-ß (TGF-ß) plays a significant role in the formation of different cancer subtypes. There is evidence that TGF-ß pathways promote cancerogenic cell characteristics but also have tumor-suppressor capabilities. The tyrosine kinase inhibitors nilotinib, dasatinib, erlotinib, gefitinib, and everolimus are approved as targeted therapies for several tumor entities, including head and neck squamous cell carcinoma (HNSCC). This study aimed to investigate the effects of these substances on the expression levels of TGFß1 and TGF-ß receptor type 2 (TGFßR2) in HPV-negative and HPV-positive SCC cell cultures. MATERIALS AND METHODS: Expression patterns of TGFß1 and TGFßR2 were determined using enzyme-linked immunosorbent assay (ELISA) in three HNSCC cell lines (i.e., HNSCC-11A, HNSCC-14C, and CERV196). These cells were incubated with nilotinib, dasatinib, erlotinib, gefitinib, and everolimus (20 µmol/l) and compared to a chemonaive control. An assessment of concentration levels was conducted after 24, 48, 72, and 96 h of treatment. RESULTS: Statistically significant changes in the expression levels of TGFß1 and TGFßR2 were found in all tested cell cultures (p<0.05) compared to the negative control. An increase in TGFß-R2 expression was detected after treatment with most of the tested tyrosine kinase inhibitors, whereas a reduction in TGFß1 was observed. The addition of everolimus had the opposite effect on both TGFßR2 and TGF-B1- expression. CONCLUSION: Expression of TGFß1 and TGFßR2 was detected in all cultured HNSCC cell lines. Nilotinib, dasatinib, erlotinib, gefitinib, and everolimus had an impact on the expression levels of TGFß1 and TGFßR2 in vitro.


Subject(s)
Dasatinib , Everolimus , Protein Kinase Inhibitors , Receptor, Transforming Growth Factor-beta Type II , Transforming Growth Factor beta1 , Humans , Everolimus/pharmacology , Transforming Growth Factor beta1/metabolism , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Dasatinib/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Gefitinib/pharmacology , Erlotinib Hydrochloride/pharmacology , Pyrimidines/pharmacology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Antineoplastic Agents/pharmacology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
3.
Int J Med Sci ; 21(8): 1461-1471, 2024.
Article in English | MEDLINE | ID: mdl-38903922

ABSTRACT

Dasatinib is one of the second-generation tyrosine kinase inhibitors used to treat chronic myeloid leukemia and has a broad target spectrum, including KIT, PDGFR, and SRC family kinases. Due to its broad drug spectrum, dasatinib has been reported at the basic research level to improve athletic performance by eliminating senescent cell removal and to have an effect on muscle diseases such as Duchenne muscular dystrophy, but its effect on myoblasts has not been investigated. In this study, we evaluated the effects of dasatinib on skeletal muscle both under normal conditions and in the regenerating state. Dasatinib suppressed the proliferation and promoted the fusion of C2C12 myoblasts. During muscle regeneration, dasatinib increased the gene expressions of myogenic-related genes (Myod, Myog, and Mymx), and caused abnormally thin muscle fibers on the CTX-induced muscle injury mouse model. From these results, dasatinib changes the closely regulated gene expression pattern of myogenic regulatory factors during muscle differentiation and disrupts normal muscle regeneration. Our data suggest that when using dasatinib, its effects on skeletal muscle should be considered, particularly at regenerating stages.


Subject(s)
Cell Differentiation , Dasatinib , Muscle Development , Muscle, Skeletal , Myoblasts , Regeneration , Dasatinib/pharmacology , Animals , Mice , Regeneration/drug effects , Cell Differentiation/drug effects , Muscle Development/drug effects , Muscle Development/genetics , Muscle, Skeletal/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/cytology , Cell Proliferation/drug effects , Humans , Cell Line , Protein Kinase Inhibitors/pharmacology
4.
Am J Physiol Cell Physiol ; 327(1): C184-C192, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38826137

ABSTRACT

Clinical experience with tyrosine kinase inhibitors (TKIs) over the past two decades has shown that, despite the apparent therapeutic benefit, nearly 30% of patients with chronic myelogenous leukemia (CML) display primary resistance or intolerance to TKIs, and approximately 25% of those treated are forced to switch TKIs at least once during therapy due to acquired resistance. Safe and effective treatment modalities targeting leukemic clones that escape TKI therapy could hence be game changers in the professional management of these patients. Here, we aimed to investigate the efficacy of a novel therapeutic oligonucleotide of unconventional design, called ASP210, to reduce BCR-ABL1 mRNA levels in TKI-resistant CML cells, with the assumption of inducing their apoptosis. Imatinib- and dasatinib-resistant sublines of BCR-ABL1-positive MOLM-7 and CML-T1 cells were established and exposed to 0.25 and 2.5 µM ASP210 for 10 days. RT-qPCR showed a remarkable reduction of the target mRNA level by >99% after a single application. Cell viability was monitored daily by trypan blue staining. In response to the lack of driver oncoprotein BCR-ABL1, TKI-resistant CML cells underwent apoptosis regardless of the presence of the clinically relevant T315I mutation by day 5 after redosing with ASP210. The effect was selective for cancer cells, indicating a favorable safety profile for this therapeutic modality. Furthermore, the spontaneous uptake and high intracellular concentrations of ASP210 suggest its potential to be effective at relatively low doses. The present findings suggest that ASP210 is a promising therapeutic avenue for patients with CML who fail to respond to TKI therapy.NEW & NOTEWORTHY Effective treatment modalities targeting leukemic clones that escape tyrosine kinase inhibitor (TKI) therapy could be game changers in the professional management of patients displaying primary resistance, intolerance, or acquired resistance to TKIs. Although delivering authentic innovations today is more complex than ever, we developed a highly potent and safe oligonucleotide-based modality against BCR-ABL1 mRNA named ASP210 that effectively induces cell death in BCR-ABL1-positive TKI-resistant cells while sparing BCR-ABL1-negative healthy cells.


Subject(s)
Apoptosis , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Oligonucleotides , Protein Kinase Inhibitors , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/pharmacology , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/metabolism , Cell Line, Tumor , Oligonucleotides/pharmacology , Apoptosis/drug effects , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Dasatinib/pharmacology , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
ACS Appl Mater Interfaces ; 16(21): 27177-27186, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38753304

ABSTRACT

Biocompatible nanoparticles as drug carriers can improve the therapeutic efficiency of hydrophobic drugs. However, the synthesis of biocompatible and biodegradable polymeric nanoparticles can be time-consuming and often involves toxic solvents. Here, a simple method for protein-based stable drug-loaded particles with a narrow polydispersity is introduced. In this process, lysozyme is mixed with hydrophobic drugs (curcumin, ellipticine, and dasatinib) and fructose to prepare lysozyme-based drug particles of around 150 nm in size. Fructose is mixed with the drug to generate nanoparticles that serve as templates for the lysozyme coating. The effect of lysozyme on the physicochemical properties of these nanoparticles is studied by transmission electron microscopy (TEM) and scattering techniques (e.g., dynamic light scattering (DLS) and small-angle X-ray scattering (SAXS)). We observed that lysozyme significantly stabilized the curcumin fructose particles for 7 days. Moreover, additional drugs, such as ellipticine and dasatinib, can be loaded to form dual-drug particles with narrow polydispersity and spherical morphology. The results also reveal that lysozyme dual ellipticine/dasatinib curcumin particles enhance the cytotoxicity and uptake on MCF-7 cells, RAW 264.7 cells, and U-87 MG cells due to the larger and rigid hydrophobic core. In summary, lysozyme in combination with fructose and curcumin can serve as a powerful combination to form protein-based stable particles for the delivery of hydrophobic drugs.


Subject(s)
Curcumin , Dasatinib , Drug Carriers , Ellipticines , Muramidase , Nanoparticles , Muramidase/chemistry , Muramidase/metabolism , Nanoparticles/chemistry , Curcumin/chemistry , Curcumin/pharmacology , Animals , Humans , Mice , Drug Carriers/chemistry , Dasatinib/chemistry , Dasatinib/pharmacology , Ellipticines/chemistry , Ellipticines/pharmacology , RAW 264.7 Cells , MCF-7 Cells , Particle Size , Fructose/chemistry , Hydrophobic and Hydrophilic Interactions , Cell Survival/drug effects , Cell Line, Tumor
6.
Int J Biol Macromol ; 269(Pt 1): 132074, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705320

ABSTRACT

Treatment for triple negative breast cancer (TNBC) remains a huge challenge due to the lack of targeted therapeutics and tumor heterogenicity. Cisplatin (Cis) have demonstrated favorable therapeutic response in TNBC and thus is used together with various kinase inhibitors to fight the heterogenicity of TNBC. The combination of Cis with SRC inhibitor dasatinib (DAS) has shown encouraging anti-TNBC efficacy although the additive toxicity was commonly observed. To overcome the severe side effects of this Cis involved therapy, here we co-encapsulated Cis and DAS into a self-assembled hyaluronan (HA) nanogel (designated as HA/Cis/DAS (HCD) nanogel) to afford the TNBC targeted delivery by using the 4T1 mouse model. The acquired HCD nanogel was around 181 nm in aqueous solution, demonstrating the pharmacological activities of both Cis and DAS. Taking advantages of HA's targeting capability towards CD44 that is overexpressed on many TNBC cells, the HCD could well maintain the anticancer efficacy of the Cis and DAS combination, significantly increase the maximum tolerated dose and relieve the renal toxicity in vivo. The current HCD nanogel provides a potent strategy to improve the therapeutic outcome of Cis and DAS combination and thus representing a new targeted treatment option for TNBC.


Subject(s)
Cisplatin , Dasatinib , Hyaluronic Acid , Nanogels , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Hyaluronic Acid/chemistry , Animals , Dasatinib/pharmacology , Dasatinib/chemistry , Mice , Cisplatin/pharmacology , Cisplatin/chemistry , Female , Nanogels/chemistry , Cell Line, Tumor , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Polyethyleneimine/chemistry , Mice, Inbred BALB C , Hyaluronan Receptors/metabolism
7.
J Med Chem ; 67(11): 9745-9758, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38819023

ABSTRACT

Herein, we describe the general design, synthesis, characterization, and biological activity of new multitargeting Pt(IV) prodrugs that combine antitumor cisplatin and dasatinib, a potent inhibitor of Src kinase. These prodrugs exhibit impressive antiproliferative and anti-invasive activities in tumor cell lines in both two-dimensional (2D) monolayers of cell cultures and three-dimensional (3D) spheroids. We show that the cisplatin moiety and dasatinib in the investigated Pt(IV) complexes are both involved in the mechanism of action in MCF7 breast cancer cells and act synergistically. Thus, combining dasatinib and cisplatin into one molecule, compared to using individual components in a mix, may bring several advantages, such as significantly higher activity in cancer cell lines and higher selectivity for tumor cells. Most importantly, Pt(IV)-dasatinib complexes hold significant promise for potential anticancer therapies by targeting epithelial-mesenchymal transition, thus preventing the spread and metastasis of tumors, a value unachievable by a simple combination of both individual components.


Subject(s)
Antineoplastic Agents , Cisplatin , Dasatinib , Drug Synergism , Prodrugs , Dasatinib/pharmacology , Dasatinib/chemistry , Dasatinib/chemical synthesis , Humans , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/chemical synthesis , Cisplatin/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Cell Line, Tumor , MCF-7 Cells , Drug Screening Assays, Antitumor , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/chemical synthesis
8.
Phytomedicine ; 130: 155705, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38761776

ABSTRACT

BACKGROUND: Senolytic combination of dasatinib and quercetin (DQ) is the most studied senolytics drugs used to treat various age-related diseases. However, its protective activity against diabetic kidney disease (DKD) and underlying mechanisms are uncertain. PURPOSE: To investigate the functions and potential mechanisms of the senolytics DQ on DKD. METHODS: Diabetic db/db mice were administrated DQ or transfected with over-expressed PPARα or shPPARα vector. The positive control group was administered irbesartan. Renal function and fibrotic changes in kidney tissue were tested. Single-cell RNA-seq (scRNA-seq) was conducted to analyze the differential transcriptome between the diabetic and control mice. Molecular docking simulation was used to assess the combination of DQ and potential factors. Moreover, tubular epithelial cells under high-glucose (HG) conditions were incubated with DQ and transfected with or without over-expressed PPARα/siPPARα vector. RESULTS: DQ significantly improved renal function, histopathological and fibrotic changes, alleviated lipid deposition, and increased ATP levels in mice with DKD. DQ reduced multiple fatty acid oxidation (FAO) pathway-related proteins and up-regulated PPARα in db/db mice. Overexpression of PPARα upregulated the expression of PPARα-targeting downstream FAO pathway-related proteins, restored renal function, and inhibited renal fibrosis in vitro and in vivo. Moreover, molecular docking and dynamics simulation analyses indicated the nephroprotective effect of DQ via binding to PPARα. Knockdown of PPARα reversed the effect of DQ on the FAO pathway and impaired the protective effect of DQ during DKD. CONCLUSION: For the first time, DQ was found to exert a renal protective effect by binding to PPARα and attenuating renal damage through the promotion of FAO in DKD.


Subject(s)
Dasatinib , Diabetic Nephropathies , Molecular Docking Simulation , PPAR alpha , Quercetin , Animals , Diabetic Nephropathies/drug therapy , Quercetin/pharmacology , PPAR alpha/metabolism , Mice , Dasatinib/pharmacology , Male , Kidney/drug effects , Kidney/pathology , Mice, Inbred C57BL , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications
9.
CPT Pharmacometrics Syst Pharmacol ; 13(7): 1144-1159, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38693610

ABSTRACT

Dasatinib, a second-generation tyrosine kinase inhibitor, is approved for treating chronic myeloid and acute lymphoblastic leukemia. As a sensitive cytochrome P450 (CYP) 3A4 substrate and weak base with strong pH-sensitive solubility, dasatinib is susceptible to enzyme-mediated drug-drug interactions (DDIs) with CYP3A4 perpetrators and pH-dependent DDIs with acid-reducing agents. This work aimed to develop a whole-body physiologically-based pharmacokinetic (PBPK) model of dasatinib to describe and predict enzyme-mediated and pH-dependent DDIs, to evaluate the impact of strong and moderate CYP3A4 inhibitors and inducers on dasatinib exposure and to support optimized dasatinib dosing. Overall, 63 plasma profiles from perorally administered dasatinib in healthy volunteers and cancer patients were used for model development. The model accurately described and predicted plasma profiles with geometric mean fold errors (GMFEs) for area under the concentration-time curve from the first to the last timepoint of measurement (AUClast) and maximum plasma concentration (Cmax) of 1.27 and 1.29, respectively. Regarding the DDI studies used for model development, all (8/8) predicted AUClast and Cmax ratios were within twofold of observed ratios. Application of the PBPK model for dose adaptations within various DDIs revealed dasatinib dose reductions of 50%-80% for strong and 0%-70% for moderate CYP3A4 inhibitors and a 2.3-3.1-fold increase of the daily dasatinib dose for CYP3A4 inducers to match the exposure of dasatinib administered alone. The developed model can be further employed to personalize dasatinib therapy, thereby help coping with clinical challenges resulting from DDIs and patient-related factors, such as elevated gastric pH.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Dasatinib , Drug Interactions , Models, Biological , Protein Kinase Inhibitors , Dasatinib/pharmacokinetics , Dasatinib/administration & dosage , Dasatinib/pharmacology , Humans , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A/metabolism , Male , Adult , Area Under Curve , Female , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Middle Aged
10.
Eur J Pharmacol ; 974: 176631, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38692425

ABSTRACT

OBJECTIVE: Dasatinib and quercetin (D & Q) have demonstrated promise in improving aged-related pathophysiological dysfunctions in humans and mice. Herein we aimed to ascertain whether the heat stress (HS)-induced cognitive deficits in aged or even young adult male mice can be reduced by D & Q therapy. METHODS: Before the onset of HS, animals were pre-treated with D & Q or placebo for 3 consecutive days every 2 weeks over a 10-week period. Cognitive function, intestinal barrier permeability, and blood-brain barrier permeability were assessed. RESULTS: Compared to the non-HS young adult male mice, the HS young adult male mice or the aged male mice had significantly lesser extents of the exacerbated stress reactions, intestinal barrier disruption, endotoxemia, systemic inflammation and oxidative stress, blood-brain barrier disruption, hippocampal inflammation and oxidative stress, and cognitive deficits evaluated at 7 days post-HS. All the cognitive deficits and other syndromes that occurred in young adult HS mice or in aged HS mice were significantly attenuated by D & Q therapy (P < 0.01). Compared to the young adult HS mice, the aged HS mice had significantly (P < 0.01) higher severity of cognitive deficits and other related syndromes. CONCLUSIONS: First, our data show that aged male mice are more vulnerable to HS-induced cognitive deficits than those of the young adult male mice. Second, we demonstrate that a combination of D and Q therapy attenuates cognitive deficits in heat stressed aged or young adult male mice via broad normalization of the brain-gut-endotoxin axis function.


Subject(s)
Blood-Brain Barrier , Dasatinib , Oxidative Stress , Quercetin , Animals , Male , Dasatinib/pharmacology , Dasatinib/therapeutic use , Quercetin/pharmacology , Quercetin/therapeutic use , Mice , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Oxidative Stress/drug effects , Aging/drug effects , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Heat-Shock Response/drug effects , Permeability/drug effects , Drug Therapy, Combination , Hippocampus/drug effects , Hippocampus/metabolism , Cognition/drug effects
11.
Pharmacol Rep ; 76(4): 793-806, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38739359

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the most difficult to treat tumors. The Src (sarcoma) inhibitor dasatinib (DASA) has shown promising efficacy in preclinical studies of PDAC. However, clinical confirmation could not be achieved. Overall, our aim was to deliver arguments for the possible reinitiating clinical testing of this compound in a biomarker-stratifying therapy trial for PDAC patients. We tested if the nanofunctionalization of DASA can increase the drug efficacy and whether certain Src members can function as clinical predictive biomarkers. METHODS: Methods include manufacturing of poly(vinyl alcohol) stabilized gold nanoparticles and their drug loading, dynamic light scattering, transmission electron microscopy, thermogravimetric analysis, Zeta potential measurement, sterile human cell culture, cell growth quantification, accessing and evaluating transcriptome and clinical data from molecular tumor dataset TCGA, as well as various statistical analyses. RESULTS: We generated homo-dispersed nanofunctionalized DASA as an AuNP@PVA-DASA conjugate. The composite did not enhance the anti-growth effect of DASA on PDAC cell lines. The cell model with high LYN expression showed the strongest response to the therapy. We confirm deregulated Src kinetome activity as a prevalent feature of PDAC by revealing mRNA levels associated with higher malignancy grade of tumors. BLK (B lymphocyte kinase) expression predicts shorter overall survival of diabetic PDAC patients. CONCLUSIONS: Nanofunctionalization of DASA needs further improvement to overcome the therapy resistance of PDAC. LYN mRNA is augmented in tumors with higher malignancy and can serve as a predictive biomarker for the therapy resistance of PDAC cells against DASA. Studying the biological roles of BLK might help to identify underlying molecular mechanisms associated with PDAC in diabetic patients.


Subject(s)
Carcinoma, Pancreatic Ductal , Dasatinib , Drug Resistance, Neoplasm , Metal Nanoparticles , Pancreatic Neoplasms , src-Family Kinases , Dasatinib/pharmacology , Dasatinib/administration & dosage , Humans , src-Family Kinases/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Gold/chemistry , Cell Proliferation/drug effects
12.
Sci Transl Med ; 16(747): eadj7685, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38748774

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is an aggressive bile duct malignancy that frequently exhibits isocitrate dehydrogenase (IDH1/IDH2) mutations. Mutant IDH (IDHm) ICC is dependent on SRC kinase for growth and survival and is hypersensitive to inhibition by dasatinib, but the molecular mechanism underlying this sensitivity is unclear. We found that dasatinib reduced p70 S6 kinase (S6K) and ribosomal protein S6 (S6), leading to substantial reductions in cell size and de novo protein synthesis. Using an unbiased phosphoproteomic screen, we identified membrane-associated guanylate kinase, WW, and PDZ domain containing 1 (MAGI1) as an SRC substrate in IDHm ICC. Biochemical and functional assays further showed that SRC inhibits a latent tumor-suppressing function of the MAGI1-protein phosphatase 2A (PP2A) complex to activate S6K/S6 signaling in IDHm ICC. Inhibiting SRC led to activation and increased access of PP2A to dephosphorylate S6K, resulting in cell death. Evidence from patient tissue and cell line models revealed that both intrinsic and extrinsic resistance to dasatinib is due to increased phospho-S6 (pS6). To block pS6, we paired dasatinib with the S6K/AKT inhibitor M2698, which led to a marked reduction in pS6 in IDHm ICC cell lines and patient-derived organoids in vitro and substantial growth inhibition in ICC patient-derived xenografts in vivo. Together, these results elucidated the mechanism of action of dasatinib in IDHm ICC, revealed a signaling complex regulating S6K phosphorylation independent of mTOR, suggested markers for dasatinib sensitivity, and described a combination therapy for IDHm ICC that may be actionable in the clinic.


Subject(s)
Adaptor Proteins, Signal Transducing , Cholangiocarcinoma , Dasatinib , Isocitrate Dehydrogenase , Mutation , src-Family Kinases , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/drug therapy , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/genetics , Dasatinib/pharmacology , Isocitrate Dehydrogenase/metabolism , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , src-Family Kinases/metabolism , src-Family Kinases/antagonists & inhibitors , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism
13.
Nat Neurosci ; 27(6): 1125-1136, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38710875

ABSTRACT

Cortical malformations such as focal cortical dysplasia type II (FCDII) are associated with pediatric drug-resistant epilepsy that necessitates neurosurgery. FCDII results from somatic mosaicism due to post-zygotic mutations in genes of the PI3K-AKT-mTOR pathway, which produce a subset of dysmorphic cells clustered within healthy brain tissue. Here we show a correlation between epileptiform activity in acute cortical slices obtained from human surgical FCDII brain tissues and the density of dysmorphic neurons. We uncovered multiple signatures of cellular senescence in these pathological cells, including p53/p16 expression, SASP expression and senescence-associated ß-galactosidase activity. We also show that administration of senolytic drugs (dasatinib/quercetin) decreases the load of senescent cells and reduces seizure frequency in an MtorS2215F FCDII preclinical mouse model, providing proof of concept that senotherapy may be a useful approach to control seizures. These findings pave the way for therapeutic strategies selectively targeting mutated senescent cells in FCDII brain tissue.


Subject(s)
Seizures , TOR Serine-Threonine Kinases , Animals , TOR Serine-Threonine Kinases/metabolism , Mice , Humans , Seizures/drug therapy , Senotherapeutics/pharmacology , Cellular Senescence/drug effects , Dasatinib/pharmacology , Epilepsy/drug therapy , Male , Malformations of Cortical Development/drug therapy , Neurons/drug effects , Neurons/metabolism , Female
14.
Blood Adv ; 8(11): 2846-2860, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38598725

ABSTRACT

ABSTRACT: The t(1;19) translocation, encoding the oncogenic fusion protein E2A (TCF3)-PBX1, is involved in acute lymphoblastic leukemia (ALL) and associated with a pre-B-cell receptor (preBCR+) phenotype. Relapse in patients with E2A-PBX1+ ALL frequently occurs in the central nervous system (CNS). Therefore, there is a medical need for the identification of CNS active regimens for the treatment of E2A-PBX1+/preBCR+ ALL. Using unbiased short hairpin RNA (shRNA) library screening approaches, we identified Bruton tyrosine kinase (BTK) as a key gene involved in both proliferation and dasatinib sensitivity of E2A-PBX1+/preBCR+ ALL. Depletion of BTK by shRNAs resulted in decreased proliferation of dasatinib-treated E2A-PBX1+/preBCR+ cells compared with control-transduced cells. Moreover, the combination of dasatinib with BTK inhibitors (BTKi; ibrutinib, acalabrutinib, or zanubrutinib) significantly decreased E2A-PBX1+/preBCR+ human and murine cell proliferation, reduced phospholipase C gamma 2 (PLCG2) and BTK phosphorylation and total protein levels and increased disease-free survival of mice in secondary transplantation assays, particularly reducing CNS-leukemic infiltration. Hence, dasatinib with ibrutinib reduced pPLCG2 and pBTK in primary ALL patient samples, including E2A-PBX1+ ALLs. In summary, genetic depletion and pharmacological inhibition of BTK increase dasatinib effects in human and mouse with E2A-PBX1+/preBCR+ ALL across most of performed assays, with the combination of dasatinib and BTKi proving effective in reducing CNS infiltration of E2A-PBX1+/preBCR+ ALL cells in vivo.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Dasatinib , Protein Kinase Inhibitors , Dasatinib/therapeutic use , Dasatinib/pharmacology , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/metabolism , Humans , Animals , Mice , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Central Nervous System Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects
15.
Biomed Pharmacother ; 175: 116606, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670048

ABSTRACT

Stress-induced premature senescent (SIPS) cells induced by various stresses deteriorate cell functions. Dasatinib and quercetin senolytics (DQ) can alleviate several diseases by eliminating senescent cells. α-tricalcium phosphate (α-TCP) is a widely used therapeutic approach for bone restoration but induces bone formation for a comparatively long time. Furthermore, bone infection exacerbates the detrimental prognosis of bone formation during material implant surgery due to oral cavity bacteria and unintentional contamination. It is essential to mitigate the inhibitory effects on bone formation during surgical procedures. Little is known that DQ improves bone formation in Lipopolysaccharide (LPS)-contaminated implants and its intrinsic mechanisms in the study of maxillofacial bone defects. This study aims to investigate whether the administration of DQ ameliorates the impairments on bone repair inflammation and contamination by eliminating SIPS cells. α-TCP and LPS-contaminated α-TCP were implanted into Sprague-Dawley rat calvaria bone defects. Simultaneously, bone formation in the bone defects was investigated with or without the oral administration of DQ. Micro-computed tomography and hematoxylin-eosin staining showed that senolytics significantly enhanced bone formation at the defect site. Histology and immunofluorescence staining revealed that the levels of p21- and p16-positive senescent cells, inflammation, macrophages, reactive oxygen species, and tartrate-resistant acid phosphatase-positive cells declined after administering DQ. DQ could partially alleviate the production of senescent markers and senescence-associated secretory phenotypes in vitro. This study indicates that LPS-contaminated α-TCP-based biomaterials can induce cellular senescence and hamper bone regeneration. Senolytics have significant therapeutic potential in reducing the adverse osteogenic effects of biomaterial-related infections and improving bone formation capacity.


Subject(s)
Bone Regeneration , Cellular Senescence , Inflammation , Osteogenesis , Rats, Sprague-Dawley , Senotherapeutics , Signal Transduction , Animals , Bone Regeneration/drug effects , Cellular Senescence/drug effects , Senotherapeutics/pharmacology , Signal Transduction/drug effects , Inflammation/drug therapy , Inflammation/pathology , Osteogenesis/drug effects , Rats , Male , Quercetin/pharmacology , Dasatinib/pharmacology , Lipopolysaccharides , Skull/drug effects , Skull/pathology
16.
Arch Toxicol ; 98(7): 2131-2142, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38589558

ABSTRACT

Oxaliplatin (OHP) is effective in colorectal cancer treatment but induces peripheral neurotoxicity (OHP-induced peripheral neurotoxicity, OIPN), diminishing survivor quality of life. Organic cation transporter 2 (OCT2) is a key OHP uptake pathway in dorsal root ganglia. Competing for OCT2-mediated OHP uptake, such as with the tyrosine kinase inhibitor dasatinib, may mitigate OHP side effects. We investigated OHP and dasatinib interaction with OCT2 in human embryonic kidney 293 (HEK293) cells expressing OCT2 within a 10-3 to 10-7 M concentration range. Uptake competition experiments using fluorescent organic cation 4-(4-dimethylaminostyryl)-N-methylpyridinium (ASP+, 1 µM) and mass spectrometry (MS) to determine cellular platinum content indicated that OHP (100 µM) is an OCT2 substrate, mediating OHP cellular toxicity. ASP+ and MS analysis revealed dasatinib as a non-transported inhibitor of hOCT2 (IC50 = 5.9 µM) and as a regulator of OCT2 activity. Dasatinib reduced transporter Vmax, potentially via Y544 phosphorylation suppression. MS analysis showed cellular dasatinib accumulation independent of hOCT2. Although 3 µM dasatinib reduced 100 µM OHP accumulation in hOCT2-HEK293 cells, co-incubation with dasatinib and OHP did not prevent OHP toxicity, possibly due to dasatinib-induced cell viability reduction. In summary, this study demonstrates OHP as an OCT2 substrate and dasatinib as a non-transported inhibitor and regulator of OCT2, offering potential for OIPN mitigation.


Subject(s)
Antineoplastic Agents , Dasatinib , Organic Cation Transporter 2 , Oxaliplatin , Protein Kinase Inhibitors , Humans , Dasatinib/pharmacology , HEK293 Cells , Oxaliplatin/pharmacology , Organic Cation Transporter 2/metabolism , Organic Cation Transporter 2/antagonists & inhibitors , Antineoplastic Agents/toxicity , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/toxicity , Drug Interactions , Pyridinium Compounds/pharmacology
17.
Biogerontology ; 25(4): 691-704, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38619669

ABSTRACT

The skin's protective functions are compromised over time by both endogenous and exogenous aging. Senescence is well-documented in skin phenotypes, such as wrinkling and sagging, a consequence of the senescence-associated secretory phenotype (SASP) that involves the accumulation of senescent fibroblasts, chronic inflammation, and collagen remodeling. Although therapeutic approaches for eliminating senescent cells from the skin are available, their efficacy remains unclear. Accordingly, we aimed to examine the effects of dasatinib in combination with quercetin (D + Q) on senescent human skin fibroblasts and aging human skin. Senescence was induced in human dermal fibroblasts (HDFs) using approaches such as long-term passaging, ionizing radiation, and doxorubicin treatment. The generated senescent cells were treated with D + Q or vehicle. Additionally, a mouse-human chimera model was generated by subcutaneously transplanting whole-skin grafts of aged individuals onto nude mice. Mouse models were administered D + Q or vehicle by oral gavage for 30 days. Subsequently, skin samples were harvested and stained for senescence-associated beta-galactosidase. Senescence-associated markers were assessed by western blotting, reverse transcription-quantitative PCR and histological analyses. Herein, D + Q selectively eliminated senescent HDFs in all cellular models of induced senescence. Additionally, D + Q-treated aged human skin grafts exhibited increased collagen density and suppression of the SASP compared with control grafts. No adverse events were observed during the study period. Collectively, D + Q could ameliorate skin aging through selective elimination of senescent dermal fibroblasts and suppression of the SASP. Our findings suggest that D + Q could be developed as an effective therapeutic approach for combating skin aging.


Subject(s)
Cellular Senescence , Dasatinib , Fibroblasts , Mice, Nude , Quercetin , Rejuvenation , Skin Aging , Skin , Dasatinib/pharmacology , Quercetin/pharmacology , Quercetin/administration & dosage , Humans , Animals , Cellular Senescence/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Skin Aging/drug effects , Skin/drug effects , Skin/metabolism , Mice , Rejuvenation/physiology , Cells, Cultured , Senescence-Associated Secretory Phenotype , Female
18.
Gynecol Oncol ; 186: 42-52, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38582027

ABSTRACT

BACKGROUND: Low grade serous ovarian carcinoma (LGSOC) is a distinct histotype of ovarian cancer characterised high levels of intrinsic chemoresistance, highlighting the urgent need for new treatments. High throughput screening in clinically-informative cell-based models represents an attractive strategy for identifying candidate treatment options for prioritisation in clinical studies. METHODS: We performed a high throughput drug screen of 1610 agents across a panel of 6 LGSOC cell lines (3 RAS/RAF-mutant, 3 RAS/RAF-wildtype) to identify novel candidate therapeutic approaches. Validation comprised dose-response analysis across 9 LGSOC models and 5 high grade serous comparator lines. RESULTS: 16 hits of 1610 screened compounds were prioritised for validation based on >50% reduction in nuclei counts in over half of screened cell lines at 1000 nM concentration. 11 compounds passed validation, and the four agents of greatest interest (dasatinib, tyrosine kinase inhibitor; disulfiram, aldehyde dehydrogenase inhibitor; carfilzomib, proteasome inhibitor; romidepsin, histone deacetylase inhibitor) underwent synergy profiling with the recently approved MEK inhibitor trametinib. Disulfiram demonstrated excellent selectivity for LGSOC versus high grade serous ovarian carcinoma comparator lines (P = 0.003 for IC50 comparison), while the tyrosine kinase inhibitor dasatinib demonstrated favourable synergy with trametinib across multiple LGSOC models (maximum zero interaction potency synergy score 46.9). The novel, highly selective Src family kinase (SFK) inhibitor NXP900 demonstrated a similar trametinib synergy profile to dasatinib, suggesting that SFK inhibition is the likely driver of synergy. CONCLUSION: Dasatinib and other SFK inhibitors represent novel candidate treatments for LGSOC and demonstrate synergy with trametinib. Disulfiram represents an additional treatment strategy worthy of investigation.


Subject(s)
Cystadenocarcinoma, Serous , Dasatinib , Drug Synergism , High-Throughput Screening Assays , Ovarian Neoplasms , Pyridones , Pyrimidinones , Humans , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Pyridones/pharmacology , Pyridones/administration & dosage , Pyrimidinones/pharmacology , Pyrimidinones/administration & dosage , Cell Line, Tumor , Dasatinib/pharmacology , Dasatinib/administration & dosage , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasm Grading , Protein Kinase Inhibitors/pharmacology , Disulfiram/pharmacology , Drug Screening Assays, Antitumor
19.
Mol Hum Reprod ; 30(5)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38603629

ABSTRACT

Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in women of reproductive age, but its pathology has not been fully characterized and the optimal treatment strategy remains unclear. Cellular senescence is a permanent state of cell-cycle arrest that can be induced by multiple stresses. Senescent cells contribute to the pathogenesis of various diseases, owing to an alteration in secretory profile, termed 'senescence-associated secretory phenotype' (SASP), including with respect to pro-inflammatory cytokines. Senolytics, a class of drugs that selectively eliminate senescent cells, are now being used clinically, and a combination of dasatinib and quercetin (DQ) has been extensively used as a senolytic. We aimed to investigate whether cellular senescence is involved in the pathology of PCOS and whether DQ treatment has beneficial effects in patients with PCOS. We obtained ovaries from patients with or without PCOS, and established a mouse model of PCOS by injecting dehydroepiandrosterone. The expression of the senescence markers p16INK4a, p21, p53, γH2AX, and senescence-associated ß-galactosidase and the SASP-related factor interleukin-6 was significantly higher in the ovaries of patients with PCOS and PCOS mice than in controls. To evaluate the effects of hyperandrogenism and DQ on cellular senescence in vitro, we stimulated cultured human granulosa cells (GCs) with testosterone and treated them with DQ. The expression of markers of senescence and a SASP-related factor was increased by testosterone, and DQ reduced this increase. DQ reduced the expression of markers of senescence and a SASP-related factor in the ovaries of PCOS mice and improved their morphology. These results indicate that cellular senescence occurs in PCOS. Hyperandrogenism causes cellular senescence in GCs in PCOS, and senolytic treatment reduces the accumulation of senescent GCs and improves ovarian morphology under hyperandrogenism. Thus, DQ might represent a novel therapy for PCOS.


Subject(s)
Cellular Senescence , Granulosa Cells , Polycystic Ovary Syndrome , Quercetin , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Female , Cellular Senescence/drug effects , Humans , Animals , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Granulosa Cells/pathology , Quercetin/pharmacology , Mice , Senescence-Associated Secretory Phenotype , Adult , Dasatinib/pharmacology , Disease Models, Animal , Senotherapeutics/pharmacology , Hyperandrogenism/pathology , Hyperandrogenism/metabolism , Interleukin-6/metabolism , Dehydroepiandrosterone/pharmacology
20.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(5): 444-452, 2024 May 09.
Article in Chinese | MEDLINE | ID: mdl-38636998

ABSTRACT

Objective: To investigate the impact of intermittent senescent cell clearance on the proliferation and differentiation of dental pulp stem cells (DPSC) in long-term, large-scale expansion, and to explore strategies for maintaining the youthful state of DPSC in vitro. Methods: Human-derived dental pulp stem cells were isolated from healthy permanent teeth extracted for orthodontic or impeding eruption reasons, provided by the Department of Oral and Maxillofacial Surgery at West China Hospital of Stomatology, Sichuan University. Long-term, large-scale in vitro expansion of DPSC was conducted. The study compared young DPSC (passage 5) with aged DPSC (passage 25) using cellular senescence-associated ß-galactosidase staining, colony formation assay, and Alizarin Red S staining for osteogenic differentiation induction. To assess the differences between the two cell populations in terms of senescence and amplification and differentiation ability. Medicine screening for the most effective senolytic was compared among 5 common senolytics [Navitoclax (ABT-263), curcumin, dasatinib, fisetin, and quercetin]. The clearance efficacy was compared using cellular senescence-associated ß-galactosidase staining to reflect the changes in senescent cell ratio. The senolytic with the highest efficacy was chosen for further experiments. The passage at which the proportion of senescent cells significantly increased was identified, and the selected senolytic was administered three times at three-generation intervals from that passage to remove senescent cells. Both the control and senolytic-treated groups were estimated by fluorescence cellular senescence-associated ß-galactosidase staining, real-time fluorescence quantitative PCR (RT-qPCR), colony formation assay, wound healing assay, and Alizarin Red S staining for osteogenic differentiation induction. Subcutaneous heterotopic osteogenesis was performed in nude mice and the grafts were analyzed by HE staining and alkaline phosphatase (ALP) immunohistochemical staining. Results: The proportion of senescent cells increased as the expansion extended, leading to decreased proliferation and osteogenic differentiation ability of senescent DPSC compared to young DPSC (P<0.05). Senescent DPSC exhibited altered mRNA expression levels of senescence-related genes, including p21, p16INK4a, IL-6, and Ki67 (P<0.001). Among the five senolytics, ABT-263 had the biggest decreases in the proportion of senescent cells. After intermittent ABT-263 treatment during expansion, the proportion of senescent cells in the senolytic-treated group [(6.72±2.34)%] was significantly lower than that in the control group [(31.82±0.57)%] (P<0.001). RT-qPCR confirmed that compared with the control group, mRNA expressions of p21, p16INK4a, and IL-6 in the senolytic-treated group were significantly decreased (P<0.05), while mRNA expressions of Ki67 were significantly increased (P<0.01). Furthermore, the cell healing ability and osteogenic differentiation ability of the senolytic-treated group were higher than those of the control group (P<0.05). In vivo experimental results indicated that the relative new bone area [(2.36±0.48)%] after DPSC transplantation in the senolytic-treated group was greater than that in the control group [(1.00±0.46)%] (P<0.05), and the expression of ALP was higher than that in the control group (P<0.01). Conclusions: ABT-263 can effectively eliminate senescent cells in long-term large-scale DPSC expansion. Continuous treatment with ABT-263 during cultivation can maintain the proliferation and differentiation ability of DPSC both in vivo and in vitro.


Subject(s)
Cell Differentiation , Cell Proliferation , Cellular Senescence , Dental Pulp , Osteogenesis , Stem Cells , Dental Pulp/cytology , Humans , Stem Cells/cytology , Osteogenesis/drug effects , Animals , Mice , Dasatinib/pharmacology , Mice, Nude , Quercetin/pharmacology , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL