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










Database
Language
Publication year range
1.
Acta Pharmacol Sin ; 41(11): 1433-1445, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32404983

ABSTRACT

In general, anti-inflammatory treatment is considered for multiple liver diseases despite the etiology. But current drugs for alleviating liver inflammation have defects, making it necessary to develop more potent and safer drugs for liver injury. In this study, we screened a series of (dihydro-)stilbene or (dihydro-)phenanthrene derivatives extracted from Pholidota chinensis for their potential biological activities. Among 31 compounds, the dihydro-stilbene gigantol exerted most potent protective effects on human hepatocytes against lithocholic acid toxicity, and exhibited solid antioxidative and anti-inflammatory effect in vitro. In mice with CCl4-induced acute liver injury, pre-administration of gigantol (10, 20, 40 mg· kg-1· d-1, po, for 7 days) dose-dependently decreased serum transaminase levels and improved pathological changes in liver tissues. The elevated lipid peroxidation and inflammatory responses in the livers were also significantly alleviated by gigantol. The pharmacokinetic studies showed that gigantol was highly concentrated in the mouse livers, which consisted with its efficacy in preventing liver injury. Using a label-free quantitative proteomic analysis we revealed that gigantol mainly regulated the immune system process in liver tissues of CCl4-treated mice, and the complement and coagulation cascades was the predominant pathway; gigantol markedly inhibited the expression of complement component C9, which was a key component for the formation of terminal complement complex (TCC) C5b-9. These results were validated by immunohistochemistry (IHC) or real time-PCR. Confocal microscopy analysis showed that gigantol significantly inhibited the vascular deposition of TCC in the liver. In conclusion, we demonstrate for the first time that oral administration of gigantol potently relieves liver oxidative stress and inflammation, possibly via a novel mechanism of inhibiting the C5b-9 formation in the liver.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Bibenzyls/therapeutic use , Guaiacol/analogs & derivatives , Inflammation/drug therapy , Oxidative Stress/drug effects , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Bibenzyls/administration & dosage , Bibenzyls/pharmacokinetics , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/pathology , Complement Membrane Attack Complex/antagonists & inhibitors , Guaiacol/administration & dosage , Guaiacol/pharmacokinetics , Guaiacol/therapeutic use , Hepatocytes/drug effects , Humans , Inflammation/pathology , Lipid Peroxidation/drug effects , Lithocholic Acid , Liver/pathology , Male , Mice, Inbred ICR , Phenanthrenes/pharmacology , Phenanthrenes/therapeutic use , Proteome/metabolism , Rats, Sprague-Dawley , Stilbenes/pharmacology , Stilbenes/therapeutic use
2.
Acta Pharmacol Sin ; 41(1): 129-137, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31341258

ABSTRACT

Rheumatoid arthritis patients can be prescribed a combination of immunosuppressive drug leflunomide (LEF) and the antiviral drug acyclovir to reduce the high risk of infection. Acyclovir is a substrate of organic anion transporter (OAT) 1/3 and multidrug resistance-associated protein (MRP) 2. Considering the extraordinarily long half-life of LEF's active metabolite teriflunomide (TER) and the kidney injury risk of acyclovir, it is necessary to elucidate the potential impact of LEF on the disposition of acyclovir. Here we used a specific MRP inhibitor MK571 and probenecid (OAT1/3 and MRP2 inhibitor) to assess the effects of MRP2 and OAT1/3 on the pharmacokinetics and tissue distribution of acyclovir in rats. We showed that LEF and probenecid, but not MK571 significantly increased the plasma concentration of acyclovir. However, kidney and liver exposures of acyclovir were increased when coadministered with LEF, probenecid or MK571. The kidney/plasma ratio of acyclovir was increased to approximately 2-fold by LEF or probenecid, whereas it was increased to as much as 14.5-fold by MK571. Consistently, these drugs markedly decreased the urinary excretion of acyclovir. TER (0.5-100 µmol/L) dose-dependently increased the accumulation of acyclovir in MRP2-MDCK cells with an IC50 value of 4.91 µmol/L. TER (5 µmol/L) significantly inhibited the uptake of acyclovir in hOAT1/3-HEK293 cells. These results suggest that LEF/TER increased the kidney accumulation of acyclovir by inhibiting the efflux transporter MRP2, which increased its kidney/plasma ratio and renal injury risk. However, the inhibitory effects of LEF/TER on OAT1/3 reduced the tubular cells' uptake of acyclovir and increased the plasma concentration.


Subject(s)
Acyclovir/pharmacokinetics , Kidney/metabolism , Leflunomide/pharmacology , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Organic Anion Transport Protein 1/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Acyclovir/administration & dosage , Acyclovir/metabolism , Administration, Intravenous , Animals , Cells, Cultured , Crotonates/administration & dosage , Crotonates/metabolism , Crotonates/pharmacology , Dogs , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hydroxybutyrates , Leflunomide/administration & dosage , Leflunomide/metabolism , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/metabolism , Male , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Nitriles , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Probenecid/administration & dosage , Probenecid/metabolism , Probenecid/pharmacology , Propionates/administration & dosage , Propionates/metabolism , Propionates/pharmacology , Quinolines/administration & dosage , Quinolines/metabolism , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Tissue Distribution , Toluidines/administration & dosage , Toluidines/metabolism , Toluidines/pharmacology
3.
Drug Deliv ; 26(1): 1058-1067, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31735064

ABSTRACT

Realgar and (-)-Epigallocatechin-3-gallate (EGCG) are natural medicines that inhibit cancer cell growth, resulting in inhibition of formation and development of tumors. The anticancer effects of realgar and EGCG were greatly improved following formulation as nanoparticles. EGCG has received increased attention as a drug carrier. The aim of this study was to prepare a new nanomedicine, (EGCG-RNPs), in which encapsulated nano-realgar. EGCG-RNPs were prepared by coprecipitation and characterized by transmission electron microscopy (TEM), differential scanning calorimetry (DSC), particle size and zeta potential, X-ray diffraction, Fourier transform infrared spectroscopy (FTIR) and in vitro release. Furthermore, we evaluated the antiproliferative effects of EGCG-RNPs on HL-60 cells in vitro, antitumor effect by intratumoral injection of EGCG-RNPs into solid tumors derived from APL HL-60 cells in vivo. Possible mechanisms were evaluated using uptake and efflux experiments in HL-60 cells. The results showed that the average particle size and zeta potentials of EGCG-RNPs was 200.3 ± 1.23 nm and -46.8 ± 1.31 mV. Controlled release of EGCG-RNPs was sustained and continued up to 72 h in vitro. Compared with nano-realgar and EGCG + RNPs (EGCG and nano-realgar physical mixing), EGCG-RNPs significantly inhibited growth of HL-60 cells. In a solid tumor model, EGCG-RNPs decreased tumor volumes, with an inhibitory rate of 60.18% at a dose of 70 mg · kg-1. The mechanisms of antitumor improvement may correlate with the increased uptake of realgar and prolonged the retention time of realgar in HL-60 cells due to EGCG as a carrier. EGCG-RNPs could enhance anticancer therapeutic efficacy for acute promyelocytic leukemia.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catechin/analogs & derivatives , Leukemia, Promyelocytic, Acute/drug therapy , Nanoparticles/chemistry , Calorimetry, Differential Scanning/methods , Catechin/chemistry , Catechin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Carriers/chemistry , HL-60 Cells , Humans , Nanomedicine/methods , Particle Size
SELECTION OF CITATIONS
SEARCH DETAIL
...