Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Free Radic Biol Med ; 204: 108-117, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37137343

ABSTRACT

Pharmacological ascorbate (P-AscH-; high dose given intravenously) generates H2O2 that is selectively cytotoxic to cancer compared to normal cells. The RAS-RAF-ERK1/2 is a major signaling pathway in cancers carrying RAS mutations and is known to be activated by H2O2. Activated ERK1/2 also phosphorylates the GTPase dynamin-related protein (Drp1), which then stimulates mitochondrial fission. Although early generation of H2O2 leads to cytotoxicity of cancer cells, we hypothesized that sustained increases in H2O2 activate ERK-Drp1 signaling, leading to an adaptive response; inhibition of this pathway would enhance the toxicity of P-AscH-. Increases in phosphorylated ERK and Drp1 induced by P-AscH- were reversed with genetic and pharmacological inhibitors of ERK and Drp1, as well as in cells lacking functional mitochondria. P-AscH- increased Drp1 colocalization to mitochondria, decreased mitochondrial volume, increased disconnected components, and decreased mitochondrial length, suggesting an increase in mitochondrial fission 48 h after treatment with P-AscH-. P-AscH- decreased clonogenic survival; this was enhanced by genetic and pharmacological inhibition of both ERK and Drp1. In murine tumor xenografts, the combination of P-AscH- and pharmacological inhibition of Drp1 increased overall survival. These results suggest that P-AscH- induces sustained changes in mitochondria, through activation of the ERK/Drp1 signaling pathway, an adaptive response. Inhibition of this pathway enhanced the toxicity P-AscH- to cancer cells.


Subject(s)
Antineoplastic Agents , Ascorbic Acid , Mitochondria , Mitochondrial Dynamics , Animals , Humans , Mice , Antineoplastic Agents/pharmacology , Ascorbic Acid/pharmacology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Hydrogen Peroxide/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Oxidative Stress/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Survival Analysis , Female
2.
Toxicol In Vitro ; 74: 105152, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33771646

ABSTRACT

Elevated expression of YY1 is known to confer anti-apoptotic phenotype and hence is an attractive target for cancer therapeutics. In a repurpose screening, towards the identification of the inhibitors of YY1 regulated transcription in gastric cancer cells, the calcium channel blockers lercanidipine and amlodipine have been identified to inhibit YY1 more efficiently. We further probed these calcium channel blockers for their potential feature of alleviating the drug resistance in gastric cancer cells. Lercanidipine and amlodipine were found to show an enhanced effect with doxorubicin in inhibiting the growth of gastric cancer cells. While doxorubicin was identified to activate the pathways TGF-ß and ERK/MAPK, lercanidipine was found to inhibit these pathways. This being the molecular mechanism behind the identified advantage of lercanidipine and amlodipine in sensitizing gastric cancer cells to doxorubicin. In multiple cellular models from different lineages, the cells with less sensitivity to doxorubicin were found to have the inherent activation of ERK/MAPK and TGF-ß pathways. Also, we have identified that doxorubicin, in combination with any of the calcium channel blockers, could inhibit the potential of cellular proliferation and spheroid formation in gastric cancer cells. The current study shows the usefulness of lercanidipine and amlodipine for the targeted and combinatorial therapeutics of gastric cancer and specifically to improve the efficiency of doxorubicin.


Subject(s)
Amlodipine/pharmacology , Antibiotics, Antineoplastic/pharmacology , Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Doxorubicin/pharmacology , Stomach Neoplasms/drug therapy , Cell Line , Cell Survival/drug effects , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Stomach Neoplasms/genetics , Transcription, Genetic , Transcriptome/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , YY1 Transcription Factor/antagonists & inhibitors
3.
Int J Mol Sci ; 21(9)2020 May 03.
Article in English | MEDLINE | ID: mdl-32375238

ABSTRACT

Extracellular signal-regulated kinase (ERK) is a part of the mitogen-activated protein kinase (MAPK) signaling pathway which allows the transduction of various cellular signals to final effectors and regulation of elementary cellular processes. Deregulation of the MAPK signaling occurs under many pathological conditions including neurodegenerative disorders, metabolic syndromes and cancers. Targeted inhibition of individual kinases of the MAPK signaling pathway using synthetic compounds represents a promising way to effective anti-cancer therapy. Cross-talk of the MAPK signaling pathway with other proteins and signaling pathways have a crucial impact on clinical outcomes of targeted therapies and plays important role during development of drug resistance in cancers. We discuss cross-talk of the MAPK/ERK signaling pathway with other signaling pathways, in particular interplay with the Hippo/MST pathway. We demonstrate the mechanism of cell death induction shared between MAPK/ERK and Hippo/MST signaling pathways and discuss the potential of combination targeting of these pathways in the development of more effective anti-cancer therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Extracellular Signal-Regulated MAP Kinases/metabolism , Neoplasms/metabolism , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hippo Signaling Pathway , Humans , Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction
4.
Molecules ; 24(21)2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31671605

ABSTRACT

Two isoforms of extracellular regulated kinase (ERK), namely ERK-1 and ERK-2, are associated with several cellular processes, the aberration of which leads to cancer. The ERK-1/2 inhibitors are thus considered as potential agents for cancer therapy. Multitarget quantitative structure-activity relationship (mt-QSAR) models based on the Box-Jenkins approach were developed with a dataset containing 6400 ERK inhibitors assayed under different experimental conditions. The first mt-QSAR linear model was built with linear discriminant analysis (LDA) and provided information regarding the structural requirements for better activity. This linear model was also utilised for a fragment analysis to estimate the contributions of ring fragments towards ERK inhibition. Then, the random forest (RF) technique was employed to produce highly predictive non-linear mt-QSAR models, which were used for screening the Asinex kinase library and identify the most potential virtual hits. The fragment analysis results justified the selection of the hits retrieved through such virtual screening. The latter were subsequently subjected to molecular docking and molecular dynamics simulations to understand their possible interactions with ERK enzymes. The present work, which utilises in-silico techniques such as multitarget chemometric modelling, fragment analysis, virtual screening, molecular docking and dynamics, may provide important guidelines to facilitate the discovery of novel ERK inhibitors.


Subject(s)
Antineoplastic Agents/analysis , Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Databases, Chemical , Discriminant Analysis , Extracellular Signal-Regulated MAP Kinases/metabolism , Ligands , Molecular Docking Simulation , Nonlinear Dynamics , Quantitative Structure-Activity Relationship , ROC Curve , Thermodynamics
5.
EBioMedicine ; 49: 106-117, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31668570

ABSTRACT

BACKGROUND: KRAS mutations are the most frequent oncogenic aberration in lung adenocarcinoma. KRAS mutant isoforms differentially shape tumour biology and influence drug responses. This heterogeneity challenges the development of effective therapies for patients with KRAS-driven non-small cell lung cancer (NSCLC). METHODS: We developed an integrative pharmacogenomics analysis to identify potential drug targets to overcome MEK/ERK inhibitor resistance in lung cancer cell lines with KRAS(G12C) mutation (n = 12). We validated our predictive in silico results with in vitro models using gene knockdown, pharmacological target inhibition and reporter assays. FINDINGS: Our computational analysis identifies casein kinase 2A1 (CSNK2A1) as a mediator of MEK/ERK inhibitor resistance in KRAS(G12C) mutant lung cancer cells. CSNK2A1 knockdown reduces cell proliferation, inhibits Wnt/ß-catenin signalling and increases the anti-proliferative effect of MEK inhibition selectively in KRAS(G12C) mutant lung cancer cells. The specific CK2-inhibitor silmitasertib phenocopies the CSNK2A1 knockdown effect and sensitizes KRAS(G12C) mutant cells to MEK inhibition. INTERPRETATION: Our study supports the importance of accurate patient stratification and rational drug combinations to gain benefit from MEK inhibition in patients with KRAS mutant NSCLC. We develop a genotype-based strategy that identifies CK2 as a promising co-target in KRAS(G12C) mutant NSCLC by using available pharmacogenomics gene expression datasets. This approach is applicable to other oncogene driven cancers. FUND: This work was supported by grants from the National Natural Science Foundation of China, the National Key Research and Development Program of China, the Lung Cancer Research Foundation and a Mildred-Scheel postdoctoral fellowship from the German Cancer Aid Foundation.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Molecular Targeted Therapy , Mutation/genetics , Pharmacogenetics , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, Dominant , Humans , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Metastasis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction
6.
Drug Des Devel Ther ; 13: 2759-2768, 2019.
Article in English | MEDLINE | ID: mdl-31496652

ABSTRACT

BACKGROUND: Various and opposite roles of epigallocatechin gallate (EGCG) have been reported in different studies. We aimed to investigate how EGCG affects the cerebral injury in a cardiac arrest/cardiopulmonary resuscitation (CA/CPR) model of rat. METHODS: The rats which were subjected to CA/CPR randomly received low dose of EGCG (3 mg/kg, Low-EGCG group, n=16), high dose of EGCG (9 mg/kg, High-EGCG group, n=16) and equal volume of 0.9% saline solution (NS group, n=16) at the first minute after return of spontaneous circulation (ROSC). The rats underwent anesthesia and intubation were defined as Sham group (n=16). Twenty-four hours after ROSC, neural defect score (NDS), ROS fluorescence intensity, degree of mitochondrial permeability transition pore (mPTP) opening, ATP contents and mitochondrial ATP synthase expression were evaluated in the four groups. The expression of extracellular signal-regulated kinase (ERK) activity and cleaved-caspase 3 were also detected by Western blot. RESULTS: CA/CPR induced severe ischemia-reperfusion injury (IRI), resulted in mitochondrial dysfunction and upregulated phosphorylation of ERK. EGCG dose-dependently alleviated the IRI after CA/CPR, inhibited ERK activity and restored mitochondrial function and, as indicated by improved NDS, reduced ROS level, decreased mPTP opening, elevated ATP content, increased ATPase expression and downregulated cleaved-caspase 3 level. CONCLUSION: EGCG alleviated global cerebral IRI by restoring mitochondrial dysfunction and ERK modulation in a rat CA/CPR model, which might make it a potential candidate agent against IRI after CA/CPR in the future. Further study is needed to determine whether higher dosage of EGCG might aggravate cerebral IRI post-CA/CPR.


Subject(s)
Cardiopulmonary Resuscitation , Catechin/analogs & derivatives , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Catechin/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart Arrest/drug therapy , Male , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
7.
Environ Toxicol ; 34(8): 968-978, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31077554

ABSTRACT

The aim of this study was to investigate the protective effects of Nano-Se against Ni-induced testosterone synthesis disorder in rats and determine the underlying protective mechanism. Sprague-Dawley rats were co-treated with Ni (5.0 mg/kg, i.p.) and Nano-Se (0.5, 1.0, and 2.0 mg/kg, oral gavage) for 14 days after which various endpoints were evaluated. The Ni-induced abnormal pathological changes and elevated 8-OHdG levels in the testes were attenuated by Nano-Se administration. Importantly, decreased serum testosterone levels in the Ni-treated rats were significantly restored by Nano-Se treatment, particularly at 1.0 and 2.0 mg/kg. Furthermore, the mRNA and protein levels of testosterone synthetase were increased by Nano-Se compared to the Ni group, whereas phosphorylated protein expression levels of mitogen-activated protein kinase (MAPK) pathways were suppressed by Nano-Se administration in the Ni-treated rats. Overall, the results suggest that Nano-Se may ameliorate the Ni-induced testosterone synthesis disturbance via the inhibition of ERK1/2, p38, and JNK MAPK pathways.


Subject(s)
MAP Kinase Signaling System/drug effects , Nickel/toxicity , Selenium/pharmacology , Testosterone/biosynthesis , Animals , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Nanoparticles , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Testis/drug effects , Testis/metabolism , Testis/pathology , Testosterone/blood , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Cell Biochem ; 120(9): 14936-14945, 2019 09.
Article in English | MEDLINE | ID: mdl-31009108

ABSTRACT

The present study was to investigate the inhibitory effect of methyl helicterate (MH) on hepatic stellate cells (HSC-T6), primarily elucidating the underlying mechanism of MH against liver fibrosis. HSC-T6 cells were activated by platelet-derived growth factor (PDGF) stimulation, and then the effects of MH on cell viability, cytomembrane integrity, colony, migration, apoptosis, and cell cycle were detected. Moreover, the regulative mechanism of MH on HSCs was investigated by detecting the activation of the extracellular signal-regulated kinase (ERK1/2) signaling pathway. The results showed that MH significantly inhibited HSC-T6 cell viability and proliferation in a concentration-dependent manner. It notably promoted the release of lactate dehydrogenase, destroying cell membrane integrity. MH also markedly inhibited HSC-T6 cell clonogenicity and migration. Moreover, MH treatment significantly induced cell apoptosis and arrested cell cycle at the G2 phase. The further study showed that MH inhibited the expression of ERK1, ERK2, c-fos, c-myc, and Ets-1, blocking the ERK1/2 pathway. In conclusion, this study demonstrates that MH significantly inhibits HSC activation and promotes cell apoptosis via downregulation of the ERK1/2 signaling pathway.


Subject(s)
Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/drug therapy , Triterpenes/pharmacology , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Drugs, Chinese Herbal/pharmacology , Enzyme Activation/drug effects , Extracellular Matrix/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , L-Lactate Dehydrogenase/metabolism , Liver Cirrhosis/pathology , MAP Kinase Signaling System/drug effects , Platelet-Derived Growth Factor/pharmacology , Rats
9.
Mediators Inflamm ; 2019: 6085801, 2019.
Article in English | MEDLINE | ID: mdl-30918469

ABSTRACT

IL-37 is an immunomodulatory cytokine that suppresses inflammation in various cell types and disease models. However, its role in keratinocytes has not been clearly understood, and there has been no report on the agents that can increase the expression of IL-37 in keratinocytes. In this study, we investigated the effects of silencing IL37 in HaCaT keratinocytes and the molecular mechanisms involved in the upregulation of IL-37 by PG102, a water-soluble extract from Actinidia arguta. It was found that knockdown of IL37 resulted in the augmented expression of antimicrobial peptides (AMPs) in response to cytokine stimulation. PG102 increased the expression of IL-37 at both mRNA and protein levels presumably by enhancing the phosphorylation of Smad3, ERK, and p38. Indeed, when cells were treated with specific inhibitors for these signaling molecules, the expression level of IL-37 was reduced. PG102 also promoted colocalization of phospho-Smad3 and IL-37. Our results suggest that IL-37 inhibits the expression of AMPs and that PG102 upregulates IL-37 through p38, ERK, and Smad3 pathways in HaCaT cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-1/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Plant Extracts/pharmacology , Smad3 Protein/metabolism , Butadienes/pharmacology , Cell Line , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Imidazoles/pharmacology , Isoquinolines/pharmacology , Nitriles/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Up-Regulation
10.
Phytomedicine ; 55: 1-8, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30668419

ABSTRACT

BACKGROUND: Many bone-related diseases such as osteoporosis and rheumatoid arthritis are commonly associated with excessive activity of the osteoclast. Ganomycin I (GMI), a meroterpenoid isolated from Vietnamese mushroom Ganoderma lucidum, possesses a variety of beneficial effects on human health. However, its impact and underlying mechanism on osteoclastogenesis remain unclear. In the present study, we investigated the effect of GMI on RANKL-induced osteoclast formation in mouse BMMs and RAW264.7 cells. METHODS: BMMs or RAW264.7 cells were treated with GMI followed by an evaluation of cell viability, RANKL-induced osteoclast differentiation, actin-ring formation, and resorption pits activity. Effects of GMI on RANKL-induced phosphorylation of MAPKs as well as the expression levels of NFATc1 and c-Fos were evaluated by Western blot analysis. Expression levels of osteoclast marker genes were evaluated by Western blot analysis and reverse transcription-qPCR. RESULTS: GMI significantly inhibited RANKL-induced osteoclast differentiation by decreasing the number of osteoclasts, osteoclast actin-ring formation, and bone resorption in a dose-dependent manner without affecting cell viability. At molecular level, GMI inhibited the RANKL-induced phosphorylation of ERK, JNK, and p38 MAPKs, as well as the expression levels of c-Fos and NFATc1, which are known to be crucial transcription factors for osteoclast formation. In addition, GMI decreased expression levels of osteoclastogenesis specific marker genes including c-Src, CtsK, TRAP, MMP-9, OSCAR, and DC-STAMP in RANKL-stimulated BMMs. CONCLUSION: Our findings suggest that GMI can attenuate osteoclast formation by suppressing RANKL-mediated MAPKs and NFATc1 signaling pathways and the anti-osteoclastogenic activity of GMI may extend our understanding of molecular mechanisms underlying biological activities and pharmacological use of G. lucidum as a traditional anti-osteoporotic medicine.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hydroquinones/pharmacology , NFATC Transcription Factors/antagonists & inhibitors , Osteogenesis/drug effects , RANK Ligand/metabolism , Animals , Bone Resorption/drug therapy , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Hydroquinones/administration & dosage , Male , Mice , Mice, Inbred ICR , NFATC Transcription Factors/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/physiology , Osteogenesis/physiology , Phosphorylation/drug effects , RAW 264.7 Cells , Reishi/chemistry
11.
J Nat Prod ; 82(2): 375-385, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30653318

ABSTRACT

During chronic inflammation, neutrophils acting locally as effector cells not only activate antibacterial defense but also promote the inflammatory response. Interleukin 8 (IL-8), the main cytokine produced by activated neutrophils, positively correlates with the severity of respiratory tract diseases. By screening European plants traditionally used for treating respiratory tract diseases, we found that extracts of aerial parts of Eupatorium cannabinum inhibit IL-8 release from neutrophils. Using bioassay-guided fractionation, we identified five sesquiterpene lactones, eupatoriopicrin (1), 5'-deoxyeupatoriopicrin (2), hiyodorilactone A (3), 3-hydroxy-5'- O-acetyleupatoriopicrin = hiyodorilactone D (4), and hiyodorilactone B (5), that efficiently (IC50 < 1 µM) inhibited IL-8 and TNF-α release in lipopolysaccharide (LPS)-stimulated human neutrophils. Moreover, all these sesquiterpene lactones suppressed the adhesion of human neutrophils to an endothelial monolayer by downregulating the expression of the ß2 integrin CD11b/CD18 on the neutrophil surface. Furthermore, eupatoriopicrin efficiently suppressed LPS-induced phosphorylation of p38 MAPK and ERK and attenuated neutrophil infiltration in the thioglycolate-induced peritonitis model in mice. Altogether, these results demonstrate the potential of the sesquiterpene lactone eupatoriopicrin as a lead substance for targeting inflammation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Interleukin-8/antagonists & inhibitors , Neutrophils/drug effects , Sesquiterpenes/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Apoptosis/drug effects , CD18 Antigens/antagonists & inhibitors , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Interleukin-8/biosynthesis , Neutrophils/physiology , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Sesquiterpenes/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
12.
Int Immunopharmacol ; 66: 236-241, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30481683

ABSTRACT

Many studies confirmed that the over-activation of RAF-MEK-ERK signaling pathway plays a central role in human cancers. To avoid drug resistance during cancer treatment, many researchers focused on the study of the downstream therapeutic target of RAF-MEK-ERK signaling pathway. Therefore, ERK1/2 became a hot anticancer target. It has been shown that ERK phosphorylation could activate Th17 cells and therefore induce inflammatory diseases. Due to these results, inhibition of ERK, as a potential drug target, could provide a solution for autoimmune diseases, especially T cell mediated diseases. In this study, a small synthetic molecule JSI287 was found with the function of alleviating IMQ-induced mice skin lesions through ERK/IL-17 signaling pathway during the screening of small molecule databases targeting ERK. The results showed that JS1287 small molecule alleviated epidermal thickness, epidermis congestion, edema and inflammatory cell infiltration, decreased release of inflammatory cytokines of IL-6, IL-12 and IL-17A, and further regulated the mRNA expression of ATF1 and protein expression of ERK1/2 in IMQ-induced skin lesions. Our study suggested that ERK inhibitor JSI287 could be a promising candidate for psoriasis treatment.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Interleukin-17/metabolism , Protein Kinase Inhibitors/pharmacology , Psoriasis/drug therapy , Signal Transduction/drug effects , Skin/drug effects , Th17 Cells/immunology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Imiquimod/toxicity , Interleukin-12/metabolism , Interleukin-6/metabolism , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred BALB C , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Psoriasis/chemically induced , Skin/pathology , Small Molecule Libraries , raf Kinases/metabolism
13.
Environ Toxicol ; 33(10): 1061-1068, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30098273

ABSTRACT

Osteoarthritis (OA) commonly affects the synovial joint and is characterized by degradation of articular cartilage. Increased matrix metalloproteinase (MMP) activity plays a major role in this degradation. Dextrose (D-glucose) prolotherapy has shown promising activity in the treatment of different musculoskeletal disorders, including OA. However, little is known about the role of glucose on MMP inhibition in OA therapy. We found that stimulating chondrocytes with the proinflammatory cytokine interleukin-1ß (IL-1ß) increased the expression of MMP-1, MMP-3, and MMP-13. Glucose reduced this increase in MMP-1 expression, but had no effect upon MMP-3 or MMP-13 expression. Analyses using a focal adhesion kinase (FAK) inhibitor, MEK inhibitors (U0126 and PD98059), an ERK inhibitor, AP-1 inhibitors (curcumin and tanshinone), or siRNAs demonstrated that the FAK, MEK, ERK, and AP-1 pathways mediate IL-1ß-induced increases in MMP-1 expression. Glucose antagonized IL-1ß-promoted phosphorylation of FAK, MEK, ERK, and c-Jun. Thus, glucose decreased IL-1ß-induced MMP-1 expression through the FAK, MEK, ERK, and AP-1 signaling cascades. These findings may provide a better understanding of the mechanisms of prolotherapy on inhibiting MMP expression.


Subject(s)
Chondrocytes/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glucose/pharmacology , Interleukin-1beta/pharmacology , Matrix Metalloproteinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line , Chondrocytes/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Signal Transduction
14.
J Hepatol ; 69(5): 1057-1065, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30030148

ABSTRACT

BACKGROUND & AIMS: Treatment of liver cancer remains challenging because of a paucity of drugs that target critical dependencies. Sorafenib is a multikinase inhibitor that is approved as the standard therapy for patients with advanced hepatocellular carcinoma, but it only provides limited survival benefit. In this study we aimed to identify potential combination therapies to improve the clinical response to sorafenib. METHODS: To investigate the cause of the limited therapeutic effect of sorafenib, we performed a CRISPR-Cas9 based synthetic lethality screen to search for kinases whose knockout synergizes with sorafenib. Synergistic effects of sorafenib and selumetinib on cell apoptosis and phospho-ERK (p-ERK) were analyzed by caspase-3/7 apoptosis assay and western blot, respectively. p-ERK was measured by immunochemical analysis using a tissue microarray containing 78 liver cancer specimens. The in vivo effects of the combination were also measured in two xenograft models. RESULT: We found that suppression of ERK2 (MAPK1) sensitizes several liver cancer cell lines to sorafenib. Drugs inhibiting the MEK (MEK1/2 [MAP2K1/2]) or ERK (ERK1/2 [MAPK1/3]) kinases reverse unresponsiveness to sorafenib in vitro and in vivo in a subset of liver cancer cell lines characterized by high levels of active p-ERK, through synergistic inhibition of ERK kinase activity. CONCLUSION: Our data provide a combination strategy for treating liver cancer and suggest that tumors with high basal p-ERK levels, which are seen in approximately 30% of liver cancers, are most likely to benefit from such combinatorial treatment. LAY SUMMARY: Sorafenib is approved as the standard therapy for patients with advanced hepatocellular carcinoma, but only provides limited survival benefit. Herein, we found that inhibition of the kinase ERK2 increases the response to sorafenib in liver cancer. Our data indicate that a combination of sorafenib and a MEK inhibitor is most likely to be effective in tumors with high basal phospho-ERK levels.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Sorafenib/administration & dosage , Biomarkers , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Phosphorylation
15.
Nanomedicine ; 14(5): 1679-1693, 2018 07.
Article in English | MEDLINE | ID: mdl-29684526

ABSTRACT

Melanoma is significantly associated with mutant BRAF gene, a suitable target for siRNA-based anti-melanoma therapy. However, a tumor-specific delivery system is a major hurdle for clinical applications. Here, we developed a novel nano-carrier, FA-GNR-siBRAF for safe topical application, which consists of folic acid (FA) as the tumor-targeting moiety, golden nanorods (GNR) providing photothermal capability to kill tumor cells under laser irradiation, and siRNA specifically silencing BRAF (siBRAF). The in vitro and in vivo results revealed that FA-GNR-siBRAF displayed high transfection rates, and subsequently induced remarkable gene knockdown of BRAF, resulting in suppression of melanoma growth due to the interruption of the MEK/ERK pathway. Combinatorial photothermal effects and BRAF knockdown by FA-GNR-siBRAF effectively killed tumor cells through apoptosis, with enhanced efficiency than individual treatments. Therefore, the FA-GNR-siBRAF simultaneously induced BRAF gene silencing and photothermal effects which achieved synergistic efficacy in the treatment of melanoma, paving a new path for developing clinical treatment methods for melanoma.


Subject(s)
Folic Acid/chemistry , Gene Silencing , Hyperthermia, Induced , Melanoma, Experimental/therapy , Nanotubes/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , RNA, Small Interfering/genetics , Animals , Apoptosis , Combined Modality Therapy , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Gold/chemistry , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Mice , Mice, Inbred C57BL , Phototherapy , Proto-Oncogene Proteins B-raf/genetics , Tumor Cells, Cultured
16.
Brain Res Bull ; 140: 132-139, 2018 06.
Article in English | MEDLINE | ID: mdl-29705048

ABSTRACT

INTRODUCTION: The signaling pathways involved in the antiepileptogenic effect of low frequency electrical stimulation (LFS) have not been fully understood. In the present study the role of extracellular signal-regulated kinase (ERK) signaling cascade was investigated in mediating the inhibitory effects of LFS on kindled seizures. METHODS: Animals received kindling stimulations for seven days (the mean number of stimulation days for achieving stage 5 seizure) according to semi-rapid perforant path kindling protocol (12 stimulations per day at 10 min intervals). LFS (0.1 ms pulse duration at 1 Hz, 800 pulses) was applied at 5 min after the last kindling stimulation every day. During the kindling procedure, FR180204 (inhibitor of ERK) was daily microinjected (1 µg/µl; intracerebroventricular) immediately after the last kindling stimulation and before LFS application. The expression of activated ERK (p-ERK) in the dentate gyrus was also investigated using immunohistochemistry technique. RESULTS: Application of LFS at 5 min after the last kindling stimulation had inhibitory effect on kindling rate. FR180204 had no significant effect on seizure parameters when administered at the dose of 1 µg/µl in kindled group of animals. However, microinjection of FR180204 before LFS application reduced the inhibitory effect of LFS on seizure severity and field potential parameters (i.e. the slope of population field excitatory postsynaptic potentials and population spike amplitude) during kindling. FR180204 also blocked the preventing effects of LFS on kindling-induced increase in early (at 10-40 ms intervals) and late (at 300-1000 ms intervals) paired pulse depression. In addition, application of LFS following kindling stimulations increased the expression of p-ERK in the dentate gyrus. CONCLUSION: Obtained results showed ERK signaling pathway had important role in mediating the antiepileptogenic effect of LFS in perforant path kindling. These findings represent a promising opportunity to gain insight about LFS mechanism in epilepsy therapy.


Subject(s)
Electric Stimulation Therapy , Epilepsy/enzymology , Epilepsy/therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Animals , Epilepsy/pathology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Kindling, Neurologic , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridazines/pharmacology , Random Allocation , Rats, Wistar , Seizures/enzymology , Seizures/pathology , Seizures/therapy
17.
J Dermatol Sci ; 91(1): 97-103, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29703420

ABSTRACT

BACKGROUND: Scleroderma is caused by aberrant transforming growth factor-ß signaling. The degradation of extracellular matrix proteins is regulated by matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs). Ultraviolet (UV) radiation has been a therapy for scleroderma. 6-Formylindolo[3,2-b]carbazole (FICZ), an endogenous aryl hydrocarbon receptor (AHR) ligand, is a tryptophan metabolite generated by UV exposure. Nonetheless, whether FICZ regulates MMPs and TIMPs has not been investigated. OBJECTIVE: To elucidate the regulatory roles of FICZ in the expression of MMPs and TIMPs in normal human dermal fibroblasts (NHDFs). METHODS: Quantitative real-time polymerase chain reaction was performed to determine the expression of MMPs or TIMPs in the NHDFs treated with FICZ or UVB. The MMPs levels were measured by enzyme-linked immunosorbent assay. The actions of FICZ on MMPs were analyzed using AHR-knockdown NHDFs or selective inhibitors of mitogen-activated protein kinases (MAPKs). Microtubule-associated protein kinase (MEK) and extracellular signal-regulated kinase (ERK) phosphorylation was examined by western blotting. RESULTS: UVB increased the mRNA and protein levels of MMP1 and MMP3 in NHDFs, while FICZ upregulated those of MMP1, but not MMP3. The effects of FICZ on TIMPs were negligible. FICZ increased MMP1 expression in an AHR-dependent manner. The FICZ-induced MMP1 upregulation was ameliorated with MEK/ERK inhibitors, whereas the effects of UVB were canceled with c-Jun N-terminal kinase (JNK) and p38-MAPK as well as MEK/ERK inhibitors. FICZ-induced ERK phosphorylation is dependent on AHR. CONCLUSION: FICZ contributes to the UV-mediated anti-fibrotic effects via the AHR/MEK/ERK signal pathway in NHDFs. FICZ is a potential therapeutic agent for scleroderma.


Subject(s)
Carbazoles/metabolism , MAP Kinase Signaling System/radiation effects , Matrix Metalloproteinase 1/metabolism , Scleroderma, Systemic/therapy , Ultraviolet Therapy/methods , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Dermis/cytology , Dermis/metabolism , Dermis/radiation effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 3/metabolism , Phosphorylation , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Scleroderma, Systemic/pathology , Tissue Inhibitor of Metalloproteinases/metabolism , Tryptophan/metabolism , Up-Regulation , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Chin J Nat Med ; 16(1): 41-52, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29425589

ABSTRACT

The aims of the present study were to evaluate the effects of puerarin on angiotensin II-induced cardiac fibroblast proliferation and to explore the molecular mechanisms of action. Considering the role of H2O2 in nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation, we hypothesized that modulating catalase activity would be a potential target in regulating the redox-sensitive pathways. Our results showed that the activation of Rac1 was dependent on the levels of intracellular H2O2. Puerarin blocked the phosphorylation of extracellular regulated protein kinases (ERK)1/2, abolished activator protein (AP)-1 binding activity, and eventually attenuated cardiac fibroblast proliferation through the inhibition of H2O2-dependent Rac1 activation. Further studies revealed that angiotensin II treatment resulted in decreased catalase protein expression and enzyme activity, which was disrupted by puerarin via the upregulation of catalase protein expression at the transcriptional level and the prolonged protein degradation. These findings indicated that the anti-proliferation mechanism of puerarin was mainly through blocking angiontensin II-triggered downregulation of catalase expression and H2O2-dependent Rac1 activation.


Subject(s)
Catalase/metabolism , Cell Proliferation/drug effects , Heart/drug effects , Hydrogen Peroxide/metabolism , Isoflavones/pharmacology , Neuropeptides/metabolism , rac1 GTP-Binding Protein/metabolism , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Animals, Newborn , Catalase/genetics , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts , Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Mice , Myocardium/cytology , Myocardium/enzymology , Myocardium/metabolism , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Transcriptional Activation/drug effects
19.
Int J Oncol ; 52(1): 201-210, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29075791

ABSTRACT

Myrmecodia platytyrea Becc., a member of the Rubiaceae family, is found throughout Southeast Asia and has been traditionally used to treat cancer. However, there is limited pharmacological information on this plant. We investigated the anticancer effects of the methanol extract of Myrmecodia platytyrea Becc. leaves (MMPL) and determined the molecular mechanisms underlying the effects of MMPL on metastasis in human hepatocellular carcinoma (HCC) cells. MMPL dose-dependently inhibited cell migration and invasion in SK­Hep1 and Huh7 cells. In addition, MMPL strongly suppressed the enzymatic activity of matrix metalloproteinases (MMP­2 and MMP­9). Diminished telomerase activity by MMPL resulted in the suppression of both telomerase activity and telomerase-associated gene expression. The levels of urokinase-type plasminogen activator receptor (uPAR) expression as well as the phosphorylation levels of signal transducer and activator of transcription 3 (STAT3) and extracellular signal-regulated kinase (ERK) were also attenuated by MMPL. The above results collectively suggest that MMPL has anticancer effects in HCC and that MMPL can serve as an effective therapeutic agent for treating human liver cancer.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Magnoliopsida/chemistry , Plant Extracts/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/isolation & purification , Matrix Metalloproteinase Inhibitors/pharmacology , Methanol/chemistry , Neoplasm Invasiveness , Plant Extracts/isolation & purification , Plant Leaves/chemistry , STAT3 Transcription Factor/metabolism
20.
Oncotarget ; 8(27): 44295-44311, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28574828

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease in urgent need of newer therapeutic modalities. Majority of patients with PDAC have mutations in KRAS, which unfortunately remains an ineffectual target. Our strategy here is to target KRAS downstream effectors PI3K and mTOR. In this study, we investigated the antitumor efficacy of the novel PI3K and mTOR dual inhibitor VS-5584 in PDAC. Our data shows that PI3K/mTOR dual inhibition causes ERK activation in all tested PDAC cell lines. Although the MEK inhibitor GSK1120212 could abrogate VS-5584-induced ERK activation, it did not substantially enhance cell death in all the cell lines tested. However, combination with ERK inhibitor SCH772984 not only mitigated VS-5584-induced ERK activation but also enhanced VS-5584-induced cell death. In a xenograft model of PDAC, we observed 28% and 44% tumor inhibition for individual treatment with VS-5584 and SCH772984, respectively, while the combined treatment showed superior tumor inhibition (80%) compared to vehicle control treatment. Our findings support the clinical development of VS-5584 and ERK inhibitor combination for PDAC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Morpholines/pharmacology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, ras , Humans , Mice , Pancreatic Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL