Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
World J Clin Cases ; 11(23): 5580-5588, 2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37637698

ABSTRACT

BACKGROUND: Clinically amyopathic deramatomyositis was manifested as the various cutaneous dermatomyositis (DM) manifestations without muscle weakness. Anti-melanoma differentiation-associated gene 5 (anti-MDA5) and anti-Ro52 antibody-dual positive clinically amyopathic DM patients are at a high risk of developing rapidly progressive interstitial lung disease, and they exhibit an immensely high half-year mortality. CASE SUMMARY: We presented three patients with anti-MDA5 and anti-Ro52 antibody-dual positive DM patients and we reviewed the previous studies on the link between anti-MDA5 and anti-Ro52 antibody-dual positive DM. Although we aggressively treated these patients similarly, but they all exhibited different prognoses. We reviewed the importance of clinical cutaneous rashes as well as the pathogenesis and treatment in the dual positive anti-MDA5 and anti-Ro52 associated DM. CONCLUSION: Patients with anti-MDA5 anti-Ro52 antibody-dual positive DM should be accurately diagnosed at an early stage and should be treated aggressively, thus, the patient's prognosis can be significantly modified.

2.
Eur J Pharm Sci ; 158: 105683, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33347980

ABSTRACT

Diacylglycerol acyltransferase 1 (DGAT1) plays a pivotal role in lipid metabolism by catalyzing the committed step in triglyceride (TG) synthesis and has been considered as a potential therapeutic target of multiple metabolic diseases, including dyslipidemia, obesity and type 2 diabetes. Here we report a novel DGAT1 inhibitor, Yhhu2407, which showed a stronger DGAT1 inhibitory activity (IC50 = 18.24 ± 4.72 nM) than LCQ908 (IC50 = 78.24 ± 8.16 nM) in an enzymatic assay and led to a significant reduction in plasma TG after an acute lipid challenge in mice. Pharmacokinetic studies illustrated that Yhhu2407 displayed a low systemic, liver- and intestine-targeted distribution pattern, which is consistent with the preferential tissue expression pattern of DGAT1 and therefore might help to maximize the beneficial pharmacological effects and prevent the occurrence of side effects. Cell-based investigations demonstrated that Yhhu2407 inhibited free fatty acid (FFA)-induced TG accumulation and apolipoprotein B (ApoB)-100 secretion in HepG2 cells. In vivo study also disclosed that Yhhu2407 exerted a beneficial effect on regulating plasma TG and lipoprotein levels in rats, and effectively ameliorated high-fat diet (HFD)-induced dyslipidemia in hamsters. In conclusion, we identified Yhhu2407 as a novel DGAT1 inhibitor with potent efficacy on improving lipid metabolism in rats and HFD-fed hamsters without causing obvious adverse effects.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Lipid Metabolism/drug effects , Animals , Diacylglycerol O-Acyltransferase/metabolism , Mice , Obesity , Rats , Triglycerides
3.
Acta Pharmacol Sin ; 42(2): 264-271, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32737468

ABSTRACT

Previous studies demonstrated that prolonged exposure to elevated levels of free fatty acids (FFA), especially saturated fatty acids, could lead to pancreatic ß-cell apoptosis, which plays an important role in the progression of type 2 diabetes (T2D). Diacylglycerol acyltransferase 1 (DGAT1), an enzyme that catalyzes the final step of triglyceride (TG) synthesis, has been reported as a novel target for the treatment of multiple metabolic diseases. In this study we evaluated the potential beneficial effects of DGAT1 inhibitors on pancreatic ß-cells, and further verified their antidiabetic effects in db/db mice. We showed that DGAT1 inhibitors (4a and LCQ908) at the concentration of 1 µM significantly ameliorated palmitic acid (PA)-induced apoptosis in MIN6 pancreatic ß-cells and primary cultured mouse islets; oral administration of a DGAT1 inhibitor (4a) (100 mg/kg) for 4 weeks significantly reduced the apoptosis of pancreatic islets in db/db mice. Meanwhile, 4a administration significantly decreased fasting blood glucose and TG levels, and improved glucose tolerance and insulin tolerance in db/db mice. Furthermore, we revealed that pretreatment with 4a (1 µM) significantly alleviated PA-induced intracellular lipid accumulation, endoplasmic reticulum (ER) stress, and proinflammatory responses in MIN6 cells, which might contribute to the protective effects of DGAT1 inhibitors on pancreatic ß-cells. These findings provided a better understanding of the antidiabetic effects of DGAT1 inhibitors.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Hypoglycemic Agents/pharmacology , Animals , Apoptosis/drug effects , Blood Glucose/drug effects , Cell Line , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Endoplasmic Reticulum Stress/drug effects , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Palmitic Acid/toxicity
4.
Zhongguo Zhong Yao Za Zhi ; 43(16): 3279-3284, 2018 Aug.
Article in Chinese | MEDLINE | ID: mdl-30200730

ABSTRACT

To establish the ultra performance liquid chromatography (UPLC) fingerprint of Dandeng Tongnao Ruanjiaonang and conduct a systemic, comprehensive quality evaluation of the drug by combining with a chemical pattern recognition method. In this study, Waters UPLC ultra-high performance liquid chromatography instrument and ACQUITY UPLCHSS T3 chromatographic colum n were employed to perform the separation with acetonitrile-0.1% formic acid aqueous solution as the mobile phase for gradient elution; and the detection wavelength was set at 256 nm to establish the UPLC fingerprint of 10 batches of Dandeng Tongnao Ruanjiaonang. Then, the further quality assessment of the drug was carried out by similarity evaluation, Cluster Analysis(CA), Principal Component Analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA). Finally, 77 peaks were recognised as common peaks in the fingerprint, and 15 peaks of them were identified using standard references. The similarity value of these 10 batches of drugs was all above 0.960, indicating a relatively stable quality. But minor differences were still discovered between the batches of the drug by CA and PCA. Finally, 6 common peaks were recognised as the quality makers using OPLS-DA method. The analysis method established in this study was scientific, accurate, reliable and simple; fingerprint combined with chemical pattern recognition technique can be used to systematically and comprehensively evaluate the drug quality of Dandeng Tongnao Ruanjiaonang; what's more, it could also provide a reference for the quality control of traditional Chinese medicine and its preparations at the same time.


Subject(s)
Drugs, Chinese Herbal/standards , Quality Control , Chromatography, High Pressure Liquid , Drugs, Chinese Herbal/chemistry , Principal Component Analysis
5.
RSC Adv ; 8(20): 11061-11069, 2018 Mar 16.
Article in English | MEDLINE | ID: mdl-35541503

ABSTRACT

CXCR1 and CXCR2 are CXC chemokine receptors (CXCRs), corresponding to cytokines of the CXC chemokine family. CXCR2 was found to be 77% homologous to CXCR1. Antagonism of the chemokine receptor CXCR2 has been proposed as a new strategy for the treatment of metastatic cancer. In order to find a CXCR2 selective antagonist, a bicyclo[2.2.1]heptane containing N,N'-diarylsquaramide (compound 2e) was identified by introducing a bridge ring system into the N,N'-diarylsquaramide skeleton, and it exhibited good CXCR2 antagonistic activity (CXCR2IC50 = 48 nM) and good selectivity (CXCR1IC50/CXCR2IC50 = 60.4). Furthermore, an in vitro biological assay of compound 2e also demonstrated its good anti-cancer metastatic effect against the pancreatic cancer cell line CFPAC1. In addition, compound 2e showed an extremely high stability in simulated intestinal fluid (SIF) and simulated gastric fluid (SGF), as well as in rat and human plasma, but not in rat and human liver microsomes. In vivo pharmacokinetic studies in rats indicated that 2e has an excellent PK profile (10 mg kg-1 po, C max = 2863 ng mL-1, t 1/2 = 2.58 h). Moreover, molecular docking was further implemented to propose the preponderant configuration of compound 2e, providing important and useful guidelines for further development.

6.
Acta Pharmacol Sin ; 37(10): 1381-1390, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27264312

ABSTRACT

AIM: C/EBP homologous protein (CHOP) is a transcription factor that is activated at multiple levels during ER stress and plays an important role in ER stress-induced apoptosis. In this study we identified a novel CHOP activator, and further investigated its potential to be a therapeutic agent for human lung cancer. METHODS: HEK293-CHOP-luc reporter cells were used in high-throughput screening (HTS) to identify CHOP activators. The cytotoxicity against cancer cells in vitro was measured with MTT assay. The anticancer effects were further examined in A549 human non-small cell lung cancer xenograft mice. The mechanisms underlying CHOP activation were analyzed using luciferase assays, and the anticancer mechanisms were elucidated in A549 cells. RESULTS: From chemical libraries of 50 000 compounds, LGH00168 was identified as a CHOP activator, which showed cytotoxic activities against a panel of 9 cancer cell lines with an average IC50 value of 3.26 µmol/L. Moreover, administration of LGH00168 significantly suppressed tumor growth in A549 xenograft bearing mice. LGH00168 activated CHOP promoter via AARE1 and AP1 elements, increased DR5 expression, decreased Bcl-2 expression, and inhibited the NF-κB pathway. Treatment of A549 cells with LGH00168 (10 µmol/L) did not induce apoptosis, but lead to RIP1-dependent necroptosis, accompanied by cell swelling, plasma membrane rupture, lysosomal membrane permeabilization, MMP collapse and caspase 8 inhibition. Furthermore, LGH00168 (10 and 20 µmol/L) dose-dependently induced mito-ROS production in A549 cells, which was reversed by the ROS scavenger N-acetyl-L-cysteine (NAC, 10 mmol/L). Moreover, NAC significantly diminished LGH00168-induced CHOP activation, NF-κB inhibition and necroptosis in A549 cells. CONCLUSION: LGH00168 is a CHOP activator that inhibits A549 cell growth in vitro and lung tumor growth in vivo.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Lung Neoplasms/drug therapy , NF-kappa B/antagonists & inhibitors , Pyrazines/therapeutic use , Pyrimidines/therapeutic use , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor/drug effects , Endoplasmic Reticulum Stress/drug effects , Humans , Mice , Mice, Inbred BALB C , Necrosis , Pyrazines/pharmacology , Pyrimidines/pharmacology
7.
Acta Pharmacol Sin ; 37(6): 719-30, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26997567

ABSTRACT

With accumulating evidence suggesting that amyloid-ß (Aß) deposition is a good diagnostic biomarker for Alzheimer's disease (AD), the discovery of active Aß probes has become an active area of research. Among the existing imaging methods, optical imaging targeting Aß aggregates (fibrils or oligomers), especially using near-infrared (NIR) fluorescent probes, is increasingly recognized as a promising approach for the early diagnosis of AD due to its real time detection, low cost, lack of radioactive exposure and high-resolution. In the past decade, a variety of fluorescent probes have been developed and tested for efficiency in vitro, and several probes have shown efficacy in AD transgenic mice. This review classifies these representative probes based on their chemical structures and functional modes (dominant solvent-dependent mode and a novel solvent-independent mode). Moreover, the pharmaceutical characteristics of these representative probes are summarized and discussed. This review provides important perspectives for the future development of novel NIR Aß diagnostic probes.


Subject(s)
Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/analysis , Fluorescent Dyes/chemistry , Optical Imaging/methods , Alzheimer Disease/pathology , Animals , Benzothiazoles , Boron Compounds/chemistry , Curcumin/chemistry , Humans , Stilbenes/chemistry , Thiazoles/chemistry , Thiophenes/chemistry
8.
Biochem Biophys Res Commun ; 466(4): 740-7, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26417688

ABSTRACT

G protein-coupled receptor 40 (GPR40) is predominantly expressed in pancreatic ß-cells and activated by long-chain fatty acids. GPR40 has drawn considerable interest as a potential therapeutic target for type 2 diabetes mellitus (T2DM) due to its important role in enhancing glucose-stimulated insulin secretion (GSIS). Encouragingly, GPR40 is also proven to be highly expressed in glucagon-like peptide-1 (GLP-1)-producing enteroendocrine cells afterwards, which opens a potential role of GPR40 in enhancing GLP-1 secretion to exert additional anti-diabetic efficacy. In the present study, we discovered a novel GPR40 agonist, yhhu4488, which is structurally different from other reported GPR40 agonists. Yhhu4488 showed potent agonist activity with EC50 of 49.96 nM, 70.83 nM and 58.68 nM in HEK293 cells stably expressing human, rat and mouse GPR40, respectively. Yhhu4488 stimulated GLP-1 secretion from fetal rat intestinal cells (FRIC) via triggering endogenous calcium store mobilization and extracellular calcium influx. The effect of yhhu4488 on GLP-1 secretion was further confirmed in type 2 diabetic db/db mice. Yhhu4488 exhibited satisfactory potency in in vivo studies. Single administration of yhhu4488 improved glucose tolerance in SD rats. Chronic administration of yhhu4488 effectively decreased fasting blood glucose level, improved ß-cell function and lipid homeostasis in type 2 diabetic ob/ob mice. Taken together, yhhu4488 is a novel GPR40 agonist that enhances GLP-1 secretion, improves metabolic control and ß-cell function, suggesting its promising potential for the treatment of type 2 diabetes.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Hypoglycemic Agents/pharmacology , Phenylpropionates/pharmacology , Pyridines/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Female , Glucose Tolerance Test , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Mutant Strains , Pregnancy , Rats , Rats, Sprague-Dawley
9.
J Am Chem Soc ; 137(38): 12296-303, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26352914

ABSTRACT

Accumulating evidence suggests that formation of peroxynitrite (ONOO(-)) in the cerebral vasculature contributes to the progression of ischemic damage, while the underlying molecular mechanisms remain elusive. To fully understand ONOO(-) biology, efficient tools that can realize the real-time tracing of endogenous ONOO(-) fluxes are indispensable. While a few ONOO(-) fluorescent probes have been reported, direct visualization of ONOO(-) fluxes in the cerebral vasculature of live mice remains a challenge. Herein, we present a fluorescent switch-on probe (NP3) for ONOO(-) imaging. NP3 exhibits good specificity, fast response, and high sensitivity toward ONOO(-) both in vitro and in vivo. Moreover, NP3 is two-photon excitable and readily blood-brain barrier penetrable. These desired photophysical and pharmacokinetic properties endow NP3 with the capability to monitor brain vascular ONOO(-) generation after injury with excellent temporal and spatial resolution. As a proof of concept, NP3 has enabled the direct visualization of neurovascular ONOO(-) formation in ischemia progression in live mouse brain by use of two-photon laser scanning microscopy. Due to these favorable properties, NP3 holds great promise for visualizing endogenous peroxynitrite fluxes in a variety of pathophysiological progressions in vitro and in vivo.


Subject(s)
Cerebrovascular Trauma/metabolism , Endothelial Cells/metabolism , Fluorescent Dyes/chemistry , Peroxynitrous Acid/metabolism , Animals , Cerebrovascular Trauma/pathology , Endothelial Cells/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/pharmacokinetics , Mice , Molecular Structure , Peroxynitrous Acid/chemistry
10.
Antimicrob Agents Chemother ; 59(11): 7061-72, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26349829

ABSTRACT

Here we first identified a novel pyridazinone derivative, compound 3711, as a nonnucleosidic hepatitis B virus (HBV) inhibitor in a cell model system. 3711 decreased extracellular HBV DNA levels by 50% (50% inhibitory concentration [IC50]) at 1.5 ± 0.2 µM and intracellular DNA levels at 1.9 ± 0.1 µM, which demonstrated antiviral activity at levels far below those associated with toxicity. Both the 3TC/ETV dually resistant L180M/M204I mutant and the adefovir (ADV)-resistant A181T/N236T mutant were as susceptible to 3711 as wild-type HBV. 3711 treatment induced the formation of genome-free capsids, a portion of which migrated faster on 1.8% native agarose gel. The induced genome-free capsids sedimented more slowly in isopycnic CsCl gradient centrifugation without significant morphological changes. 3711 treatment decreased levels of HBV DNA contained in both secreted enveloped virion and naked virus particles in supernatant. 3711 could interfere with capsid formation of the core protein (Cp) assembly domain. A Cp V124W mutant, which strengthens capsid interdimer interactions, recapitulated the effect of 3711 on capsid assembly. Pyridazinone derivative 3711, a novel chemical entity and HBV inhibitor, may provide a new opportunity to combat chronic HBV infection.


Subject(s)
Antiviral Agents/pharmacology , Capsid/metabolism , Hepatitis B virus/drug effects , Virus Replication/drug effects , Capsid Proteins/metabolism , DNA, Viral/genetics , Drug Resistance, Viral
11.
Acta Pharmacol Sin ; 36(3): 343-52, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25732571

ABSTRACT

AIM: Defects in fatty acid metabolism contribute to the pathogenesis of insulin resistance and obesity. In this study, we investigated the effects of a novel compound yhhu981 on fatty acid metabolism in vitro and in vivo. METHODS: The capacity to stimulate fatty acid oxidation was assessed in C2C12 myotubes. The fatty acid synthesis was studied in HepG2 cells using isotope tracing. The phosphorylation of AMPK and acetyl-CoA carboxylase (ACC) was examined with Western blot analysis. For in vivo experiments, ob/ob mice were orally treated with yhhu981 acutely (300 mg/kg) or chronically (150 or 300 mg·kg(-1)·d(-1) for 22 d). On the last day of treatment, serum and tissue samples were collected for analysis. RESULTS: Yhhu981 (12.5-25 µmol/L) significantly increased fatty acid oxidation and the expression of related genes (Sirt1, Pgc1α and Mcad) in C2C12 myotubes, and inhibited fatty acid synthesis in HepG2 cells. Furthermore, yhhu981 dose-dependently increased the phosphorylation of AMPK and ACC in both C2C12 myotubes and HepG2 cells. Compound C, an AMPK inhibitor, blocked fatty acid oxidation in yhhu981-treated C2C12 myotubes and fatty acid synthesis decrease in yhhu981-treated HepG2 cells. Acute administration of yhhu981 decreased the respiratory exchange ratio in ob/ob mice, whereas chronic treatment with yhhu981 ameliorated the lipid abnormalities and ectopic lipid deposition in skeletal muscle and liver of ob/ob mice. CONCLUSION: Yhhu981 is a potent compound that stimulates fatty acid oxidation, and exerts pleiotropic effects on lipid metabolism by activating AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Alkynes/pharmacology , Anti-Obesity Agents/pharmacology , Enzyme Activators/pharmacology , Fatty Acids/metabolism , Liver/drug effects , Muscle Fibers, Skeletal/drug effects , Obesity/drug therapy , Resorcinols/pharmacology , AMP-Activated Protein Kinases/antagonists & inhibitors , Acetyl-CoA Carboxylase/metabolism , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Enzyme Activation , Hep G2 Cells , Humans , Liver/enzymology , Mice, Obese , Muscle Fibers, Skeletal/enzymology , Obesity/enzymology , Oxidation-Reduction , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Up-Regulation
12.
Acta Pharmacol Sin ; 35(8): 1074-81, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25066323

ABSTRACT

AIM: To discover novel hepatitis C virus (HCV) inhibitors and elucidate the mechanism of action of the active compounds. METHODS: HCV subgenomic replicon-based luciferase reporter cell line was used to screen 1200 synthetic compounds with novel structures. Huh7.5.1 cell line stably transfected with HCV NS3/4A protease reporter was established to investigate the anti-HCV mechanism of the active compounds. The active compounds were further examined in an in vitro HCV infection assay to confirm their anti-HCV activity. RESULTS: After two-round screening in the anti-HCV replicon assay, some 2,4-diaminoquinazoline derivatives and carboxamide analogues were found to possess anti-HCV replicon activities (the IC50 values were less than 5 µmol/L). Among them, two representative compounds HZ-1157 and LZ-110618-6 inhibited HCV NS3/4A protease with IC50 values of 1.0 and 0.68 µmol/L, respectively. Furthermore, HZ-1157 and LZ-110618-6 inhibited HCV infection in vitro with IC50 values of 0.82 and 0.11 µmol/L, respectively. CONCLUSION: Some 2,4-diaminoquinazoline derivatives and carboxamide analogues have been identified as novel anti-HCV compounds.


Subject(s)
Antiviral Agents/chemistry , Hepacivirus/drug effects , Hepatitis C/drug therapy , Quinazolines/chemistry , Quinazolines/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Amides/chemistry , Amides/pharmacology , Cell Line , Drug Evaluation, Preclinical , Hepacivirus/metabolism , Hepatitis C/virology , Humans , Viral Nonstructural Proteins/metabolism
13.
Acta Pharmacol Sin ; 35(1): 89-97, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24241352

ABSTRACT

AIM: c-Met kinase deregulation is strongly associated with the formation, progression and dissemination of human cancers. In this study we identified Yhhu3813 as a small-molecule inhibitor of c-Met kinase and characterized its antitumor properties both in vitro and in vivo. METHODS: The activities of different kinases were measured using ELISA assays and signaling proteins in the cells were detected with Western blotting. Cell proliferation was assessed using SRB or MTT assay in twenty human cell lines and cell cycle distribution was determined with flow cytometry. Transwell-based assay was used to evaluate cell migration and invasion. Cell invasive growth was detected by a morphogenesis assay. c-Met overactivated human NSCLC cell line EBC-1 xenografts were used to evaluate the in vivo anti-tumor efficacy. RESULTS: Yhhu3813 potently inhibited c-Met kinase activity in vitro with an IC50 value of 2.4±0.3 nmol/L, >400-fold higher than that for a panel of 15 different tyrosine kinases, suggesting a high selectivity of Yhhu3813. The compound (20, 100 and 500 nmol/L) dose-dependently inhibited the phosphorylation of c-Met and its key downstream Akt and Erk signal cascades in multiple c-Met aberrant human cancer cell lines, regardless of the mechanistic complexity in c-Met activation across different cellular contexts. In 20 human cancer cell lines harboring different backgrounds of c-Met expression/activation, Yhhu3813 potently inhibited c-Met-driven cell proliferation via arresting cells at G1/S phase. Furthermore, Yhhu3813 substantially impaired c-Met-mediated cell migration, invasion, scattering, and invasive growth. Oral administration of EBC-1 xenograft mice with Yhhu3813 (50 or 100 mg·kg(-1)·d(-1), qd, for 2 weeks) dose-dependently suppressed the tumor growth, which was correlated with a reduction in the intratumoral proliferation index and c-Met signaling. CONCLUSION: Yhhu3813 is a potent selective inhibitor of c-Met that inhibits c-Met-dependent neoplastic phenotypes of human cancer cells in vitro and in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Phenotype , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Dogs , Dose-Response Relationship, Drug , Female , HCT116 Cells , HT29 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Protein Kinase Inhibitors/therapeutic use , Random Allocation , Xenograft Model Antitumor Assays/methods
14.
Acta Pharmacol Sin ; 33(9): 1204-16, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684031

ABSTRACT

AIM: To characterize a small molecule compound HK-156 as a novel inhibitor of the nuclear factor κB (NF-κB) signaling pathway. METHODS: THP-1 monocytes and HEK293/hTLR4A-MD2-CD14 cells were tested. HK-156 and compound 809, an HK-156 analogue, were synthesized. A luciferase assay was used to evaluate the transcriptional activity of NF-κB. The levels of cytokines were measured with cytokine arrays, ELISA and quantitative PCR. An electrophoretic mobility shift assay (EMSA), immunofluorescence, Western blot and mass spectrometry were used to investigate the molecular mechanisms underlying the actions of the agent. BALB/c mice challenged with lipopolysaccharide (LPS, 15 mg/kg, ip) were used as a mouse experimental endotoxemia model. RESULTS: In HEK293hTLR4/NF-κB-luc cells treated with LPS (1000 ng/mL), HK-156 inhibited the transcriptional activity of NF-κB in a concentration-dependent manner (IC50=6.54 ± 0.37 µmol/L). Pretreatment of THP-1 monocytes with HK-156 (5, 10 and 20 µmol/L) significantly inhibited LPS-induced release and production of TNF-α and IL-1ß, attenuated LPS-induced translocation of NF-κB into the nucleus and its binding to DNA, and suppressed LPS-induced phosphorylation and degradation of IκBα, and phosphorylation of IKKß and TGFß-activated kinase (TAK1). Meanwhile, HK-156 (5, 10 and 20 µmol/L) slightly suppressed LPS-induced activation of p38. The effect of HK-156 on LPS-induced activation of NF-κB signaling was dependent on thiol groups of cysteines in upstream proteins. In mouse models of sepsis, pre-injection of HK-156 (50 mg/kg, iv) significantly inhibited TNFα production and reduced the mortality caused by the lethal dose of LPS. CONCLUSION: HK-156 inhibits LPS-induced activation of NF-κB signaling by suppressing the phosphorylation of TAK1 in vitro, and exerts beneficial effects in a mouse sepsis model. HK-156 may therefore be a useful therapeutic agent for treating sepsis.


Subject(s)
Acrylamides/pharmacology , NF-kappa B/metabolism , Pyrimidines/pharmacology , Sepsis/drug therapy , Signal Transduction/drug effects , Acrylamides/administration & dosage , Animals , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Endotoxemia/drug therapy , Endotoxemia/physiopathology , Enzyme-Linked Immunosorbent Assay , Female , HEK293 Cells , Humans , Inhibitory Concentration 50 , Injections, Intravenous , Lipopolysaccharides , MAP Kinase Kinase Kinases , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Polymerase Chain Reaction , Pyrimidines/administration & dosage , Sepsis/physiopathology , Survival
15.
J Med Chem ; 55(7): 3135-43, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22448770

ABSTRACT

The results of a high-throughput screening assay using the DENV-2 replicon showed that the 2,4-diaminoquinazoline derivative 4a has a high dengue virus inhibitory activity (EC(50) = 0.15 µM). A series of 2,4-diaminoquinazoline derivatives based on 4a as a lead compound were synthesized and subjected to structure-antidengue activity relationship studies. Among the series of 2,4-diaminoquinazoline derivative probed, 4o was observed to display both the highest antiviral potency (EC(50) = 2.8 nM, SI > 1000) and an excellent pharmacokinetic profile.


Subject(s)
Antiviral Agents/chemical synthesis , Dengue Virus/drug effects , Quinazolines/chemical synthesis , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Cell Line , Dengue Virus/genetics , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , High-Throughput Screening Assays , Humans , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Rats , Replicon/drug effects , Structure-Activity Relationship
16.
Acta Pharmacol Sin ; 31(11): 1470-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21052084

ABSTRACT

AIM: To test whether pharmacological inhibition of Diacylglycerol acyltransferase 1 (DGAT1) by a small-molecule inhibitor H128 can improve metabolism disorders in leptin receptor-deficient db/db mice. METHODS: To investigate the effect of H128 on intestinal fat absorption,db/db mice were acutely given a bolus of corn oil by gavage. The mice were further orally administered H128 (3 and 10 mg/kg) for 5 weeks. Blood glucose, lipids, insulin, ALT, and AST as well as hepatic triglycerides were measured. The insulin tolerance test was performed to evaluate insulin sensitivity. The expression of genes involved in fatty acid oxidation was detected by RT-PCR. RESULTS: Oral administration of H128 (10 mg/kg) acutely inhibited intestinal fat absorption following a lipid challenge in db/db mice. Chronic treatment with H128 significantly inhibited body weight gain, decreased food intake, and induced a pronounced reduction of serum triglycerides. In addition, H128 treatment markedly ameliorated hepatic steatosis, characterized by decreased liver weight, lipid droplets, and triglyceride content as well as serum ALT and AST levels. Furthermore, H128 treatment increased the expression of the CPT1 and PPARα genes in liver, suggesting that H128 enhanced fatty acid oxidation in db/db mice. However, neither blood glucose nor insulin tolerance was affected by H128 treatment throughout the 5-week experimental period. CONCLUSION: DGAT1 may be an effective therapeutic target for the treatment of obesity, hyperlipidemia and hepatic steatosis.


Subject(s)
Biphenyl Compounds/pharmacology , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fatty Liver/drug therapy , Hyperlipidemias/drug therapy , Phenylurea Compounds/pharmacology , Receptors, Leptin/physiology , Weight Gain/drug effects , Animals , Biphenyl Compounds/chemistry , Biphenyl Compounds/therapeutic use , Blood Glucose/analysis , Eating/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Fatty Acids/metabolism , Fatty Liver/enzymology , Fatty Liver/metabolism , Hyperlipidemias/enzymology , Hyperlipidemias/metabolism , Insulin Resistance , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Knockout , Phenylurea Compounds/chemistry , Phenylurea Compounds/therapeutic use , Receptors, Leptin/genetics , Triglycerides/biosynthesis , Triglycerides/blood
17.
Mol Cell Biochem ; 330(1-2): 181-5, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19412730

ABSTRACT

Hepatic stellate cells (HSCs) play an important role in the development of hepatic fibrosis. Heat shock protein 90 (Hsp90) is essential for the maturation and activity of a varied group of proteins involved in signal transduction and cell cycle regulation. In this study, we found that two Hsp90 inhibitors, VER-49009 and its analog VER-49009M, inhibited the proliferation of hepatic stellate cell line CFSC cells, and both of them induced G2 phase arrest in CFSC cells. Akt expression was decreased by the treatment of Hsp90 inhibitors in CFSC cells. Based on these findings, we propose that the inhibition of Hsp90 might be a rational approach in the prevention of liver fibrosis.


Subject(s)
Cell Proliferation/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hepatic Stellate Cells/cytology , Animals , Cell Line , G2 Phase/drug effects , Hepatic Stellate Cells/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Pyrazoles/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...