Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Nat Med ; 78(2): 370-381, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38265612

ABSTRACT

The Wnt/ß-catenin signaling pathway plays important roles in several cancer cells, including cell proliferation and development. We previously succeeded in synthesizing a small molecule compound inhibiting the Wnt/ß-catenin signaling pathway, named LPD-01 (1), and 1 inhibited the growth of human colorectal cancer (HT-29) cells. In this study, we revealed that 1 inhibits the growth of HT-29 cells stronger than that of another human colorectal cancer (SW480) cells. Therefore, we have attempted to identify the target proteins of 1 in HT-29 cells. Firstly, we investigated the effect on the expression levels of the Wnt/ß-catenin signaling pathway-related proteins. As a result, 1 inhibited the expression of target proteins of Wnt/ß-catenin signaling pathway (c-Myc and Survivin) and their genes, whereas the amount of transcriptional co-activator (ß-catenin) was not decreased, suggesting that 1 inhibited the Wnt/ß-catenin signaling pathway without affecting ß-catenin. Next, we investigated the target proteins of 1 using magnetic FG beads. Chemical pull-down assay combined with mass spectrometry suggested that 1 directly binds to importin7. As expected, 1 inhibited the nuclear translocation of importin7 cargoes such as Smad2 and Smad3 in TGF-ß-stimulated HT-29 cells. In addition, the knockdown of importin7 by siRNA reduced the expression of target genes of Wnt/ß-catenin signaling pathway. These results suggest that importin7 is one of the target proteins of 1 for inhibition of the Wnt/ß-catenin signaling pathway.


Subject(s)
Colorectal Neoplasms , beta Catenin , Humans , beta Catenin/metabolism , Wnt Signaling Pathway , Cell Proliferation , Colorectal Neoplasms/drug therapy , Cell Line, Tumor
2.
Int Immunopharmacol ; 107: 108553, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35358777

ABSTRACT

Recently, baicalin refers to flavonoid compound extracted from Scutellaria baicalensis Georgi has been indicated to hold promising therapeutic effects in alcohol-associated liver disease (ALD). However, knowledge of the molecular mechanisms for its hepatoprotective effect is still very limited. Evidence exists suggesting potential association between miR-205 and baicalin's function. Bioinformatic analysis and dual luciferase reporter assay were conducted to determine the binding affinity between miR-205 and importinα5. Our findings revealed that baicalin could alleviate ALD by raising the expression of miR-205. Additionally, miR-205 repressed NF-κB signaling pathway activation by binding to importinα5 to relieve ALD. Baicalin inhibited importinα5-mediated NF-κB signaling pathway to protect the liver against alcohol-induced injury, inflammation, oxidative stress and hepatocyte apoptosis. Taken conjointly, baicalin confers hepatoprotective effect against ALD through miR-205-mediated importinα5 inhibition via the NF-κB signaling pathway, highlighting a promising therapeutic target for ALD treatment with the help of traditional Chinese medicine.


Subject(s)
Liver Diseases, Alcoholic , MicroRNAs , Flavonoids/pharmacology , Flavonoids/therapeutic use , Humans , Liver Diseases, Alcoholic/drug therapy , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism
3.
Biochem Pharmacol ; 186: 114501, 2021 04.
Article in English | MEDLINE | ID: mdl-33684389

ABSTRACT

Lactucopicrin, a bitter sesquiterpene lactone of leafy vegetables, such as chicory, curly escarole, and lettuce, possesses anti-malarial, anti-cancer and analgesic properties. However, it remains unknown whether lactucopicrin could inhibit vascular endothelial nuclear factor-κB (NF-κB) activation, a hallmark of vascular inflammatory diseases including sepsis. In tumor necrosis factor-α-stimulated human or mouse aortic endothelial cells, lactucopicrin dose-dependently inhibited NF-κB activation, and concomitantly repressed both vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1)-mediated monocyte adhesion. The lactucopicrin effect was not due to modulation of inhibitor of NF-κB kinases (IKK) α/ß/γ, inhibitor of NF-κB alpha (IκBα), and NF-κB/p65 DNA binding activity. Instead, lactucopicrin inhibited importin-α3 expression by destabilization of its mRNA, an effect mediating the lactucopicrin effect on NF-κB activity. More importantly, in lipopolysaccharide (LPS)-elicited septic mice, oral gavage with lactucopicrin decreased mortality by 30.5% as compared with the control treatment. This effect was associated with inhibited importin-α3 expression, suppressed NF-κB activation and VCAM-1/ICAM-1 expression, and inhibited leukocyte influx in the vascular endothelium of both lung and aorta. Collectively, our novel data suggest that dietary supplementation with lactucopicrin inhibits endothelial NF-κB activation by down-regulation of importin-α3 and thereby improves sepsis.


Subject(s)
Endothelial Cells/metabolism , Lactones/therapeutic use , NF-kappa B/metabolism , Sepsis/drug therapy , Sepsis/metabolism , Sesquiterpenes/therapeutic use , alpha Karyopherins/metabolism , Animals , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , HL-60 Cells , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Lactones/pharmacology , Male , Mice , Mice, Inbred C57BL , Sepsis/chemically induced , Sesquiterpenes/pharmacology , alpha Karyopherins/antagonists & inhibitors
4.
Infect Genet Evol ; 88: 104699, 2021 03.
Article in English | MEDLINE | ID: mdl-33385575

ABSTRACT

Plasmodium falciparum, the prime causative agent of malaria, is responsible for 4, 05,000 deaths per year and fatality rates are higher among the children aged below 5 years. The emerging distribution of the multi-drug resistant P. falciparum becomes a worldwide concern, so the identification of unique targets and novel inhibitors is a prime need now. In the present study, we have employed pharmacoinformatics approaches to analyze 265 lead-like compounds from PubChem databases for virtual screening. Thereafter, 15 lead-like compounds were docked within the active side pocket of importin alpha. Comparative ligand properties and absorption, distribution, metabolism, excretion, and toxicity (ADMET) profile were also assessed. Finally, a novel inhibitor was designed and assessed computationally for its efficacy. From the comparative analysis we have found that our screened compounds possess better results than the existing lead ivermectin; having the highest binding energy of -15.6 kcal/mol, whereas ivermectin has -12.4 kcal/mol. The novel lead compound possessed more fascinating output without deviating any of the rules of Lipinski. It also possessed higher bioavailability and the drug-likeness score of 0.55 and 0.71, respectively compared to ivermectin. Furthermore, the binding study was confirmed by molecular dynamics simulation over 25 ns by evaluating the stability of the complex. Finally, all the screened compounds and the novel compound showed promising ADMET properties likewise. To end, we hope that our proposed screened compounds, as well as the novel compound, might give some advances to treat malaria efficiently in vitro and in vivo.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/chemistry , Plasmodium falciparum/drug effects , alpha Karyopherins/chemistry , beta Karyopherins/chemistry , Drug Design , Drug Discovery , Drug Evaluation, Preclinical , Drug Resistance , Humans , Ligands , Malaria, Falciparum/parasitology , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , alpha Karyopherins/metabolism , beta Karyopherins/metabolism
5.
Eur J Med Chem ; 178: 636-647, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31226655

ABSTRACT

Deregulation of NF-κB plays an important role in various diseases by controlling cell growth, inflammation, the immune response, and cytokine production. Although many NF-κB inhibitors have been developed, to the best of our knowledge, none of them have been successfully translated into clinical practice as medicines. To overcome this issue, we aimed to develop a new class of NF-κB inhibitors. Previous reports indicated that the N-terminal cysteine is a promising target for NF-κB. Based on this, we first selected 10 natural products or their derivatives from the natural product library that we developed and examined the effect on NF-κB and the viability of cancer cells with constitutively strong NF-κB activity. Among them, we found that an azoxy natural product, jietacin A, with a vinylazoxy group and an aliphatic side chain, reduced cell viability and inhibited nuclear translocation of free NF-κB. In addition, we performed design, synthesis, and biological evaluation of jietacin derivatives for development of a novel NF-κB inhibitor. Of these derivatives, a fully synthesized derivative 25 with vinylazoxy and ynone groups had a potent effect. We clarified the structure-activity relationship of this compound. Jietacin A and 25 also inhibited tumor necrosis factor-α-mediated induction of NF-κB. The NF-κB inhibitory effect depended on the N-terminal cysteine and the neighboring Arg-Ser-Ala-Gly-Ser-Ile (RSAGSI) domain of NF-κB. We also found that 25 inhibited the association between NF-κB and importin α, suggesting inhibition of NF-κB at an early step of nuclear translocation. Overall, this study indicated that the vinylazoxy motif may compose a new class of NF-κB inhibitors, providing further insight for rational drug design and rendering a unique mode of action.


Subject(s)
Azo Compounds/pharmacology , Biological Products/pharmacology , Drug Discovery , NF-kappa B/antagonists & inhibitors , Azo Compounds/chemical synthesis , Azo Compounds/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Structure , NF-kappa B/metabolism , Structure-Activity Relationship
7.
Exp Ther Med ; 16(2): 1052-1060, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30112050

ABSTRACT

Compound Astragalus and Salvia miltiorrhiza extract (CASE) is a Chinese herbal formula consisting of astragalosides, astragalus polysaccharide and salvianolic acids extracted from Astragalus membranaceus and Salvia miltiorhiza. Previous studies by our group have demonstrated that CASE effectively suppresses diethylinitrosamine (DEN)-induced hepatocellular carcinoma (HCC) in rats via modulating transforming growth factor ß/Mothers against decapentaplegic (TGFß/Smad) signaling. To further elucidate the mechanism of CASE, the effects of CASE on TGF-ß1, the serine/threonine kinase receptors of TGF-ß [TGF-ß receptor type-I (TßRI) and TßRII] and karyopherins [Importin 7 (Imp7) and Imp8], which are crucial for TGF-ß/Smad signaling in fibro-hepatocarcinogenesis, were assessed in the present study using in vivo (DEN-induced HCC in rats) and in vitro [TGF-ß1-stimulated rat myofibroblasts (MFBs) and HepG2 cells] models of fibro-hepatocarcinogenesis. Hematoxylin and eosin staining revealed that CASE may suppress inflammatory reactions and fibrosis in HCC as well as increasing the differentiation of HCC cells. Positive TGF-ß1 staining was increased in HCC nodule areas and in adjacent normal liver tissues in DEN-treated rats, while TßRI staining was increased only in normal adjacent liver tissues. The elevated expression of TGF-ß1, TßRI and TßRII was suppressed by CASE. CASE treatment also reduced glutathione S-transferase P 1 and Imp7/8 protein expression in fibro-hepatocarcinogenesis. In vitro experiments confirmed that CASE was able to decrease the expression of TßRI and TßRII in TGF-ß1-stimulated MFBs and HepG2 cells. These results indicate that the anti-HCC effect of CASE may be achieved by mediating TGF-ß/TßR and Imp7/8 protein expression, suggesting that CASE has multiple targets in HCC treatment.

SELECTION OF CITATIONS
SEARCH DETAIL