Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Phytother Res ; 36(9): 3601-3618, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35871535

ABSTRACT

Puerarin, the major bioactive ingredient isolated from the root of Pueraria lobata (Willd.), attenuates body weight gain and reduces lipid levels in high-fat diet-induced obese mice; however, the underlying mechanism responsible for regulating lipid metabolism remains unclear. This study investigated the molecular mechanism(s) underlying the role of puerarin in regulating lipogenesis and lipolysis in human HepG2 cells. In this study, puerarin strongly inhibited the expression of fatty acid synthase (FASN) and sterol regulatory element binding protein 1c (SREBP-1c). Moreover, puerarin significantly induced the expression of adipose triglyceride lipase (ATGL), which is responsible for triacylglycerol hydrolase activity in cells. Puerarin enhanced 5' AMP-activated protein kinase (AMPK) activity, which is a central regulator of hepatic lipid metabolism. Furthermore, this AMPK activation could be mediated by sirtuin 1 (SIRT1) and calcium signaling pathways involved in G protein-coupled estrogen receptor (GPER) signaling. GPER blockage significantly reversed the effect of puerarin on lipid accumulation and the related signaling pathways. Docking studies showed that puerarin could bind in the GPER in a similar manner as GPER agonist G1. Our results suggest that puerarin can improve hepatic steatosis by activating GPER; it's signaling cascade sequentially induced calcium and SIRT1 signaling pathways. Thus, puerarin may be a potential therapeutic agent for the treatment of non-alcoholic fatty liver disease.


Subject(s)
Non-alcoholic Fatty Liver Disease , Sirtuin 1 , AMP-Activated Protein Kinases/metabolism , Animals , Calcium/metabolism , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/pharmacology , Hep G2 Cells , Humans , Isoflavones , Lipid Metabolism , Lipids , Liver , Mice , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Sirtuin 1/metabolism
2.
Cell Chem Biol ; 26(6): 818-829.e9, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30982749

ABSTRACT

Covalent kinase inhibitors, which typically target cysteine residues, represent an important class of clinically relevant compounds. Approximately 215 kinases are known to have potentially targetable cysteines distributed across 18 spatially distinct locations proximal to the ATP-binding pocket. However, only 40 kinases have been covalently targeted, with certain cysteine sites being the primary focus. To address this disparity, we have developed a strategy that combines the use of a multi-targeted acrylamide-modified inhibitor, SM1-71, with a suite of complementary chemoproteomic and cellular approaches to identify additional targetable cysteines. Using this single multi-targeted compound, we successfully identified 23 kinases that are amenable to covalent inhibition including MKNK2, MAP2K1/2/3/4/6/7, GAK, AAK1, BMP2K, MAP3K7, MAPKAPK5, GSK3A/B, MAPK1/3, SRC, YES1, FGFR1, ZAK (MLTK), MAP3K1, LIMK1, and RSK2. The identification of nine of these kinases previously not targeted by a covalent inhibitor increases the number of targetable kinases and highlights opportunities for covalent kinase inhibitor development.


Subject(s)
Acrylamide/pharmacology , Cysteine/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Acrylamide/chemistry , Cell Line, Tumor , Cysteine/metabolism , Drug Discovery , Humans , Ligands , Protein Kinase Inhibitors/chemistry
3.
Phytomedicine ; 24: 134-140, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-28160853

ABSTRACT

BACKGROUND: Acetylshikonin is one of the biologically active compounds derived from the root of Lithospermum erythrorhizon, a medicinal plant with anti-cancer and anti-inflammation activity. Although there have been a few previous reports demonstrating that acetylshikonin exerts anti-cancer activity in vitro and in vivo, it is still not clear what is the exact molecular target protein of acetylshikonin in cancer cells. PURPOSE: The purpose of this study is to evaluate the inhibitory effect of acetylshikonin against CYP2J2 enzyme which is predominantly expressed in human tumor tissues and carcinoma cell lines. STUDY DESIGN: The inhibitory effect of acetylshikonin on the activities of CYP2J2-mediated metabolism were investigated using human liver microsomes (HLMs), and its cytotoxicity against human hepatoma HepG2 cells was also evaluated. METHOD: Astemizole, a representative CYP2J2 probe substrate, was incubated in HLMs in the presence or absence of acetylshikonin. After incubation, the samples were analyzed by liquid chromatography and triple quadrupole mass spectrometry. The anti-cancer activity of acetylshikonin was evaluated on human hepatocellular carcinoma HepG2 cells. WST-1, cell counting, and colony formation assays were further adopted for the estimation of the growth rate of HepG2 cells treated with acetylshikonin. RESULTS: Acetylshikonin inhibited CYP2J2-mediated astemizole O-demethylation activity (Ki = 2.1µM) in a noncompetitive manner. The noncompetitive inhibitory effect of acetylshikonin on CYP2J2 enzyme was also demonstrated using this 3D structure, which showed different binding location of astemizole and acetylshikonin in CYP2J2 model. It showed cytotoxic effects against human hepatoma HepG2 cells (IC50 = 2µM). In addition, acetylshikonin treatment inhibited growth of human hepatocellular carcinoma HepG2 cells leading to apoptosis accompanied with p53, bax, and caspase3 activation as well as bcl2 down-regulation. CONCLUSION: Taken together, our present study elucidates acetylshikonin displays the inhibitory effects against CYP2J2 in HLMs and anti-cancer activity in human hepatocellular carcinoma HepG2 cells.


Subject(s)
Anthraquinones/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Cytochrome P-450 Enzyme System/metabolism , Hep G2 Cells/drug effects , Liver Neoplasms/drug therapy , Microsomes, Liver/drug effects , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Phytotherapy , Plant Extracts/therapeutic use
4.
Exp Mol Med ; 48: e229, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-27126178

ABSTRACT

p21-activated kinases (PAKs) are key regulators of actin dynamics, cell proliferation and cell survival. Deregulation of PAK activity contributes to the pathogenesis of various human diseases, including cancer and neurological disorders. Using an ELISA-based screening protocol, we identified naphtho(hydro)quinone-based small molecules that allosterically inhibit PAK activity. These molecules interfere with the interactions between the p21-binding domain (PBD) of PAK1 and Rho GTPases by binding to the PBD. Importantly, they inhibit the activity of full-length PAKs and are selective for PAK1 and PAK3 in vitro and in living cells. These compounds may potentially be useful for determining the details of the PAK signaling pathway and may also be used as lead molecules in the development of more selective and potent PAK inhibitors.


Subject(s)
Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Allosteric Regulation/drug effects , Cell Line , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Enzyme Activation/drug effects , Humans , Isoenzymes , Protein Binding , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries , Structure-Activity Relationship , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/chemistry
5.
J Ethnopharmacol ; 172: 254-60, 2015 Aug 22.
Article in English | MEDLINE | ID: mdl-26116162

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Tripterygium wilfordii (lei gong teng; Thunder of God Vine), which belongs to the Celastraceae family, has long been used in traditional Chinese medicine to treat inflammation and rheumatoid arthritis. Celastrol is a bioactive compound isolated from T. wilfordii. AIM OF THE STUDY: We investigated whether celastrol suppressed binding of lipopolysaccharides (LPS) to myeloid differentiation factor 2 (MD2), thereby downregulating Toll-like receptor4 (TLR4) activation in mouse primary macrophages. MATERIALS AND METHODS: Cytokine expression was determined by polymerase chain reaction analysis and enzyme-linked immunosorbent assay in bone marrow-derived primary macrophages (BMDMs). The kinase activity of tank-binding kinase 1 (TBK1) was examined by a luciferase reporter assay and an in vitro kinase assay. LPS binding to MD2 was examined by an in vitro binding assay and confocal microscopy analysis. RESULTS: Celastrol reduced LPS-induced expression of inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-12, and IL-1ß, at both the mRNA and protein levels in BMDMs. Celastrol suppressed LPS binding to MD2, as shown by the in vitro binding assay, whereas it did not inhibit TBK1. In addition, co-localization of LPS with MD2 in BMDMs was blocked by celastrol. The inhibitory effects of celastrol on LPS binding to MD2 were reversed by thiol donors (N-acetyl-L-cysteine and dithiothreitol), suggesting that the thiol reactivity of celastrol contributes to its inhibitory effects on TLR4 activation in macrophages. CONCLUSION: Our results demonstrate that celastrol suppresses TLR4 activation through the inhibition of LPS binding to the TLR4/MD2 complex. These results provide a novel mechanism of action by which celastrol contributes to the anti-inflammatory activity of T. wilfordii.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , Toll-Like Receptor 4/metabolism , Triterpenes/pharmacology , Acetylcysteine/pharmacology , Animals , Anti-Inflammatory Agents/isolation & purification , Cytokines/metabolism , Dithiothreitol/pharmacology , Enzyme-Linked Immunosorbent Assay , Lipopolysaccharides/metabolism , Lymphocyte Antigen 96/metabolism , Macrophages/metabolism , Medicine, Chinese Traditional , Mice , Mice, Inbred C57BL , Pentacyclic Triterpenes , Polymerase Chain Reaction , Sulfhydryl Compounds/chemistry , Tripterygium/chemistry , Triterpenes/isolation & purification
6.
Int J Oncol ; 45(2): 558-66, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24841075

ABSTRACT

Daurinol, a lignan from the ethnopharmacological plant Haplophyllum dauricum, was recently reported to be a novel topoisomerase II inhibitor and an alternative to the clinical anticancer agent etoposide based on a colorectal cancer model. In the present study, we elucidated the detailed biochemical mechanism underlying the inhibition of human topoisomerase IIα by daurinol based on a molecular docking study and in vitro biochemical experiments. The computational simulation predicted that daurinol binds to the ATP-binding pocket of topoisomerase IIα. In a biochemical assay, daurinol (10-100 µM) inhibited the catalytic activity of topo-isomerase IIα in an ATP concentration-dependent manner and suppressed the ATP hydrolysis activity of the enzyme. However, daurinol did not inhibit topoisomerase I activity, most likely because topoisomerase I does not contain an ATP-binding domain. We also evaluated the anti-proliferative activity of daurinol in ovarian, small cell lung and testicular cancer cells, common target cancers treated with etoposide. Daurinol potently inhibited SNU-840 human ovarian cancer cell proliferation through cell cycle arrest in S phase, while etoposide induced G2/M phase arrest. Daurinol induced the increased expression of cyclin E, cyclin A and E2F-1, which are important proteins regulating S phase initiation and progression. Daurinol did not induce abnormal cell and nuclear enlargement in SNU-840 cells, in contrast to etoposide. Based on these data, we suggest that daurinol is a potential anticancer drug candidate for the treatment of human ovarian cancer with few side effects.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzodioxoles/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Naphthalenes/pharmacology , Ovarian Neoplasms/pathology , S Phase/drug effects , Topoisomerase II Inhibitors/pharmacology , Antigens, Neoplasm , Blotting, Western , Cell Line, Tumor , DNA Topoisomerases, Type II , DNA-Binding Proteins/antagonists & inhibitors , Female , Humans , Molecular Docking Simulation
7.
PLoS One ; 9(3): e91508, 2014.
Article in English | MEDLINE | ID: mdl-24618722

ABSTRACT

Thiacremonone (2, 4-dihydroxy-2, 5-dimethyl-thiophene-3-one) is an antioxidant substance as a novel sulfur compound generated from High-Temperature-High-Pressure-treated garlic. Peroxiredoxin 6 (PRDX6) is a member of peroxidases, and has glutathione peroxidase and calcium-independent phospholipase A2 (iPLA2) activities. Several studies have demonstrated that PRDX6 stimulates lung cancer cell growth via an increase of glutathione peroxidase activity. A docking model study and pull down assay showed that thiacremonone completely fits on the active site (cys-47) of glutathione peroxidase of PRDX6 and interacts with PRDX6. Thus, we investigated whether thiacremonone inhibits cell growth by blocking glutathione peroxidase of PRDX6 in the human lung cancer cells, A549 and NCI-H460. Thiacremonone (0-50 µg/ml) inhibited lung cancer cell growth in a concentration dependent manner through induction of apoptotic cell death accompanied by induction of cleaved caspase-3, -8, -9, Bax, p21 and p53, but decrease of xIAP, cIAP and Bcl2 expression. Thiacremonone further inhibited glutathione peroxidase activity in lung cancer cells. However, the cell growth inhibitory effect of thiacremonone was not observed in the lung cancer cells transfected with mutant PRDX6 (C47S) and in the presence of dithiothreitol and glutathione. In an allograft in vivo model, thiacremonone (30 mg/kg) also inhibited tumor growth accompanied with the reduction of PRDX6 expression and glutathione peroxidase activity, but increased expression of cleaved caspase-3, -8, -9, Bax, p21 and p53. These data indicate that thiacremonone inhibits tumor growth via inhibition of glutathione peroxidase activity of PRDX6 through interaction. These data suggest that thiacremonone may have potentially beneficial effects in lung cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Neoplasms/metabolism , Neoplasms/pathology , Peroxiredoxin VI/genetics , Thiophenes/pharmacology , Allografts , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Garlic/chemistry , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , Mice , Models, Molecular , Molecular Conformation , Mutation , Neoplasms/drug therapy , Peroxiredoxin VI/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Protein Binding , Thiophenes/chemistry , Thiophenes/metabolism , Tumor Burden/drug effects
8.
J Antimicrob Chemother ; 67(1): 49-58, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22076990

ABSTRACT

OBJECTIVES: Hepatitis C virus (HCV) infection causes chronic liver disease and is a major public health problem worldwide. The aim of this study was to evaluate the potential of Monascus pigment derivatives, which were derived from a microbial secondary metabolite synthesized from polyketides by Monascus spp., as HCV antiviral agents. METHODS: We performed an in vitro RNA-dependent RNA polymerase (RdRp) assay to screen for HCV RdRp inhibitors. The anti-HCV activity of RdRp inhibitors in HCV-replicating cells was evaluated by quantification of the RNA viral genome. Molecular docking analysis was performed to predict the binding sites of the selected RdRp inhibitors. RESULTS: We have identified a Monascus pigment and its derivatives as inhibitors of the HCV NS5B RdRp. A group of Monascus orange pigment (MOP) amino acid derivatives, in which the reactive oxygen moiety was changed to amino acids, significantly inhibited HCV replication. Further, combination of the MOP derivatives (Phe, Val or Leu conjugates) with interferon (IFN)-α inhibited HCV replication more than IFN-α treatment alone. Lastly, molecular docking studies indicate the inhibitors may bind to a thumb subdomain allosteric site of NS5B. The antiviral activity of the MOP derivatives was related to a modulation of the mevalonate pathway, since the mevalonate-induced increase in HCV replication was suppressed by the MOP compounds. CONCLUSIONS: Our results identify amino acid derivatives of MOP as potential anti-HCV agents and suggest that their combination with IFN-α might offer an alternative strategy for the control of HCV replication.


Subject(s)
Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Hepacivirus/drug effects , Mevalonic Acid/metabolism , Monascus/chemistry , Pigments, Biological/pharmacology , Virus Replication/drug effects , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Binding Sites , Biosynthetic Pathways/genetics , DNA-Directed RNA Polymerases/chemistry , Drug Evaluation, Preclinical/methods , Humans , Molecular Dynamics Simulation , Pigments, Biological/chemistry , Pigments, Biological/isolation & purification
9.
Bioorg Med Chem Lett ; 21(11): 3329-34, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21531135

ABSTRACT

We report the use of pharmacophore-based virtual screening as an efficient tool for the discovery of novel HCV polymerase inhibitors. A three-dimensional pharmacophore model for the HCV-796 binding site, NNI site IV inhibitor, to the enzyme was built by means of the structure-based focusing module in Cerius2 program. Using these models as a query for virtual screening, we produced a successful example of using pharmacophore-based virtual screening to identify novel compounds with HCV replicon assay through inhibition of HCV polymerization. Among the hit compounds, compounds 1 and 2 showed 56% and 48% inhibition of NS5B polymerization activity at 20 µM, respectively. In addition, compound 1 also exhibited replicon activity with EC(50) value of 2.16 µM. Following up the initial hit, we obtained derivatives of compound 1 and evaluated polymerization inhibition activity and HCV replicon assay. These results provide information necessary for the development of more potent NS5B inhibitors.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical , Hepacivirus/enzymology , Humans , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL