Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Pharmacol Sin ; 43(3): 724-734, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34117368

ABSTRACT

Hepatocellular carcinoma (HCC), the most prevalent liver cancer, is considered one of the most lethal malignancies with a dismal outcome mainly due to frequent intrahepatic and distant metastasis. In the present study, we demonstrated that oroxylin A, a natural product extracted from Scutellaria radix, significantly inhibits transforming growth factor-beta1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) and metastasis in HCC. Oroxylin A blocked the TGF-ß1/Smad signaling via upregulating the non-steroidal anti-inflammatory drug-activated gene-1 (NAG-1) expression. Oroxylin A promoted NAG-1 transcription by regulating the acetylation of CCAAT/enhancer binding protein ß (C/EBPß), a transcription factor that binds to the NAG-1 promoter. In terms of the underlying mechanism, oroxylin A may interact with histone deacetylase 1 (HDAC1) by forming hydrogen bonds with GLY149 residue and induce proteasome-mediated degradation of HDAC1 subsequently impairing HDAC1-mediated deacetylation of C/EBPß and promoting the expression of NAG-1. Taken together, our findings revealed a previously unknown tumor-suppressive mechanism of oroxylin A. Oroxylin A should be further investigated as a potential clinical candidate for inhibiting HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Flavonoids/pharmacology , Growth Differentiation Factor 15/drug effects , Liver Neoplasms/pathology , CCAAT-Binding Factor/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Histone Deacetylase 1/drug effects , Humans , Smad Proteins/drug effects , Transforming Growth Factor beta1/drug effects
2.
Theranostics ; 11(1): 93-106, 2021.
Article in English | MEDLINE | ID: mdl-33391463

ABSTRACT

Rationale: Acute myeloid leukemia (AML) is a common type of haematological malignancy. Several studies have shown that neoplasia in AML is enhanced by tyrosine kinase pathways. Recently, given that aberrant activation of Fms-like tyrosine receptor kinase 3 (FLT3) acts as a critical survival signal for cancer cells in 20‒30% patients with AML, inhibition of FLT3 may be a potential therapeutic strategy. Therefore, we identified LT-171-861, a novel kinase inhibitor with remarkable inhibitory activity against FLT3, in preclinical models of AML. Methods: We determined the inhibitory effects of LT-171-861 in vitro using AML cell lines and transformed BaF3 cells. Target engagement assays were used to verify the interaction between LT-171-861 and FLT3. Finally, a subcutaneous model and a bone marrow engrafted model were used to evaluate the antitumor effects of LT­171­861 in vivo. Results: Our data demonstrated that LT-171-861 had high affinity for FLT3 protein. We also showed that LT-171-861 had an inhibitory effect on FLT3 mutants in cellular assays. Moreover, LT-171-861 had a growth-inhibitory effect on human AML cell lines harboring FLT3 internal tandem duplications (FLT3-ITD) such as FLT3-D835Y, FLT3­ITD-N676D, FLT3-ITD-D835Y, FLT3-ITD-F691L, FLT3-ITD-Y842C and AML blasts from patients with FLT3-ITD. Furthermore, LT-171-861 showed potent antileukemic efficacy against AML cells. We also show the efficacy of LT­171-861 in a subcutaneous implantation model and a bone marrow engrafted model in vivo, where administration of LT-171-861 led to almost complete tumor regression and increased survival. Conclusions: Overall, this study not only identifies LT-171-861 as a potent FLT3 inhibitor, but also provides a rationale for the upcoming clinical trial of LT-171-861 in patients with AML and FLT3-ITD mutations.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Heterocyclic Compounds/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , CSK Tyrosine-Protein Kinase/drug effects , Cell Line , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/drug effects , Female , Heterocyclic Compounds/therapeutic use , Humans , Inhibitory Concentration 50 , Janus Kinases/drug effects , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/drug effects , Purines/therapeutic use , THP-1 Cells , fms-Like Tyrosine Kinase 3/genetics
3.
Free Radic Biol Med ; 112: 49-59, 2017 11.
Article in English | MEDLINE | ID: mdl-28697922

ABSTRACT

We have previously reported that a newly synthesized compound, GL-V9 could induce mitochondria-mediated apoptosis in HepG2 cells. However, the underlying mechanisms have not been fully understood yet. In current study, we further showed that GL-V9 exhibited significant inhibitory effect on growth of several hepatocellular carcinoma cell lines. Moreover, GL-V9-induced growth inhibition was coincident with the strong upregulation of nonsteroidal anti-inflammatory drug-activated gene-1 (NAG-1), a TGFß superfamily member, which has been linked with tumor suppression. Further analysis uncovered that GL-V9-activated p38 MAPK pathway contributed to enhancement of NAG-1 mRNA stability. Interestingly, we observed that the intracellular NAG-1 protein induced by GL-V9 could, at least in part, localize in mitochondria where it might affect protein expression, thereby resulting in dissipation of mitochondria membrane potential (MMP) and accumulation of mitochondrial superoxide, eventually facilitating to apoptosis events. Silence of NAG-1 could attenuate mitochondria related apoptosis caused by GL-V9. Moreover, GL-V9 suppressed tumor growth in xenograft model accompanied with upregulation of NAG-1 in tumor tissues. Collectively, these data demonstrated that NAG-1 could play an important role in mitochondria apoptosis triggered by GL-V9, thus providing novel mechanistic explanations and potential target for using GL-V9 as a chemotherapeutic agent against human hepatocellular carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic , Growth Differentiation Factor 15/genetics , Liver Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Growth Differentiation Factor 15/agonists , Growth Differentiation Factor 15/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/agonists , Signal Transduction , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...