Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 11(7): e0160285, 2016.
Article in English | MEDLINE | ID: mdl-27467078

ABSTRACT

BACKGROUND: Cytochrome P450 2C19 (CYP2C19) is an important drug-metabolizing enzyme (DME), which is responsible for the biotransformation of several kinds of drugs such as proton pump inhibitors, platelet aggregation inhibitors and antidepressants. Previous studies showed that Buchang NaoXinTong capsules (NXT) increased the CYP2C19 metabolic activity in vitro and enhanced the antiplatelet effect of clopidogrel in vivo. However, the underlying molecular mechanism remained unclear. In the present study, we examined whether Pregnane X receptor (PXR) plays a role in NXT-mediated regulation of CYP2C19 expression. METHODS: We applied luciferase assays, real-time quantitative PCR (qPCR), Western blotting and cell-based analysis of metabolic activity experiments to investigate the NXT regulatory effects on the CYP2C19 promoter activity, the mRNA/ protein expression and the metabolic activity. RESULTS: Our results demonstrated that NXT significantly increased the CYP2C19 promoter activity when co-transfected with PXR in HepG2 cells. Mutations in PXR responsive element abolished the NXT inductive effects on the CYP2C19 promoter transcription. Additionally, NXT incubation (150 and 250µg/mL) also markedly up-regulated endogenous CYP2C19 mRNA and protein levels in PXR-transfected HepG2 cells. Correspondingly, NXT leaded to a significant enhancement of the CYP2C19 catalytic activity in PXR-transfected HepG2 cells. CONCLUSION: In summary, this is the first study to suggest that NXT could induce CYP2C19 expression via PXR activation.


Subject(s)
Cytochrome P-450 CYP2C19/metabolism , Drugs, Chinese Herbal/pharmacology , Receptors, Steroid/metabolism , Up-Regulation/drug effects , Cytochrome P-450 CYP2C19/genetics , Hep G2 Cells , Humans , Pregnane X Receptor , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Transcription, Genetic
2.
Pharmazie ; 70(10): 668-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26601424

ABSTRACT

MicroRNA-184 (miR-184) is found to be significantly deregulated in human cancers associated with tumorigenesis and progression. In this study, we aimed to investigate the role and mechanism of miR-184 expression in epithelial ovarian cancer (EOC). Relative expression of miR-184 was measured by quantificational real-time polymerase chain reaction assay (qRT-PCR) in 80 EOC patients. Kaplan-Meier curve and the log-rank test were conducted to detect the prognostic value of miR-184. Function assays including cell proliferation, apoptosis and inflammation were further explored in vitro. We found that miR-184 was down-regulated in EOC tissues and cell lines compared with paired non-cancerous tissues and IOSE, respectively. Moreover, miR-184 was expressed at significantly lower levels in late-stage (III/IV) EOC tissues. Cox regression multivariate analysis indicated that miR-184 and FIGO stage were independent prognostic indicators for EOC patients. Patients with high miR-184 level achieved significantly a higher 5-year survival rate compared with low level group (P < 0.001). Functional assays showed that miR-184 over-expression could suppress EOC cell proliferation as well as inflammation and induce apoptosis in vitro. Altogether, our results suggest that miR-184 together with pathologic diagnosis is critical for prognosis determination in EOC patients and help select treatment strategy.


Subject(s)
Apoptosis/physiology , Biomarkers, Tumor/analysis , Cell Proliferation , Inflammation/pathology , MicroRNAs/analysis , MicroRNAs/physiology , Neoplasms, Glandular and Epithelial/diagnosis , Ovarian Neoplasms/diagnosis , Apoptosis/genetics , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cytokines/metabolism , Female , Humans , Inflammation/genetics , MicroRNAs/genetics , Middle Aged , Prognosis , Transfection
3.
Pharmazie ; 70(9): 581-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26492642

ABSTRACT

Niemann-Pick C1-like 1 (NPC1L1i) protein is the key transporter responsible for dietary cholesterol absorption. Recent studies indicated that several functional polymorphisms of NPC1L1 were associated with coronary heart disease (CHD) and response to ezetimibe therapy. The aim of the present study was to analyze the allele frequency and haplotype distribution of NPC1L1 polymorphisms in Chinese Hans and to compare them with those of other ethnic populations reported before. Blood samples were collected from 424 unrelated Chinese Hans (246 males and 178 females). Ten NPC1L1 polymorphisms (-762T > C, -133A > G, -18C > A, 1721C > T, 1735C > G, 1764T > C, 1767G > A, 27677T > C, 25342A > C and 28650A > G) were genotyped by direct sequencing or polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assay. Among the variants, the minor allele frequency of -762T > C and 1735C > G were 35.0% and 37.0%, respectively. Furthermore, these two polymorphisms were highly linked with a D' value of 0.80. The observed frequencies of two major haplotypes were 59.1% for T-762/C1735 and 30.1% for C-762/G1735, respectively. The frequencies of the rest variants were extremely low (1.8% for - 133G, 1.5% for -18A, 0.9% for 1721T and only 0.2% for 27677C allele, respectively) or even not detected (1764T > C, 1767G > A, 25342A > C and 28650A > G) in our study population. Comparison with other ethnic populations revealed a remarkable genetic variability in the incidences of NPC1L1 polymorphisms. The frequencies of NPC1L1 polymorphisms in Chinese Hans are comparable to Japanese population but totally different from Caucasians, African-Americans and Hispanic individuals. This is the first study to report the ethnic difference in the frequencies of NPC1L1 functional polymorphisms in detail. -762T > C and 1735C > G are two prevalent NPC1L1 variants which need further studies to explore their clinical impact on CHD prevalence and response to ezetimibe therapy in Chinese Hans.


Subject(s)
Membrane Proteins/genetics , Adult , Alleles , Asian People , China/epidemiology , Female , Gene Frequency , Genotype , Haplotypes , Humans , Male , Membrane Transport Proteins , Middle Aged , Polymorphism, Genetic/genetics
4.
Pharmacogenomics ; 16(8): 855-63, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26086348

ABSTRACT

AIMS: To explore the effect of urate transporter 1 (URAT1) polymorphisms on the hypertensive patients with hyperuricemia and the uricosuric action of losartan therapy among hypertensive patients with hyperuricemia. METHODS: 101 hypertensive patients with hyperuricemia were detected the genotypes of URAT1 rs1529909 and rs3825016 and undergo a 2-weeks following losartan treatment. Before and after treatment, serum uric acid (SUA) and other clinical data were compared between different genotypes of URAT1 patients. RESULTS: The frequency of rs3825016 (C/T) CT genotype was significant higher in the hypertensive patients with hyperuricemia than that in the healthy controls (32.7 vs 18.8%; p = 0.02). After lorsatan treatment, the patients with the rs3825016 (C/T) or rs1529909 (T/C) mutant genotypes had lower decreased value (DV) of SUA compared with the patients who are wild-type of the variant (p = 0.001 and p < 0.001, respectively). Combined the two variants together, the DV of SUA in two variants both wild-type patients higher than that in the two variants mutant patients (p < 0.0001). CONCLUSION: These results suggest that URAT1 rs3825016 and rs1529909 polymorphisms influence the uricosuric action of losartan. Original submitted 20 August 2014; Revision submitted 15 April 2015.


Subject(s)
Hypertension/drug therapy , Hyperuricemia/drug therapy , Losartan/administration & dosage , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Aged , Female , Genetic Association Studies , Genotype , Humans , Hypertension/complications , Hypertension/genetics , Hyperuricemia/complications , Hyperuricemia/genetics , Losartan/adverse effects , Male , Middle Aged , Polymorphism, Single Nucleotide , Uricosuric Agents/administration & dosage , Uricosuric Agents/adverse effects
5.
PLoS One ; 9(9): e106540, 2014.
Article in English | MEDLINE | ID: mdl-25247304

ABSTRACT

Lectin-like low-density lipoprotein receptor 1 (LOX-1) is a receptor for oxidized low density lipoprotein (oxLDL) in endothelial cells. The activation of LOX-1 by oxLDL stimulates the apoptosis and dysfunction of endothelial cells, and contributes to atherogenesis. However, the regulatory factors for LOX-1 are still unclear. MicroRNAs are small, endogenous, non-coding RNAs that regulate gene expressions at a post-transcriptional level. The let-7 family is the second microRNA been discovered, which plays important roles in cardiovascular diseases. Let-7a and let-7b were predicted to target LOX-1 3'-UTR and be highly expressed in endothelial cells. The present study demonstrated that LOX-1 was a target of let-7a and let-7b. They inhibited the expression of LOX-1 by targeting the positions of 310-316 in LOX-1 3'-UTR. Over-expression of let-7a and let-7b inhibited the oxLDL-induced endothelial cell apoptosis, NO deficiency, ROS over-production, LOX-1 upregulation and endothelial nitric oxide synthase (eNOS) downregulation. Moreover, we found that oxLDL treatment induced p38MAPK phosphorylation, NF-κB nuclear translocation, IκB degradation and PKB dephosphorylation. Let-7a or let-7b over-expression attenuated these alterations significantly. The present study may provide a new insight into the protective properties of let-7a and let-7b in preventing the endothelial dysfunction associated with cardiovascular disease, such as atherosclerosis.


Subject(s)
Endothelial Cells/drug effects , Lipoproteins, LDL/pharmacology , MicroRNAs/metabolism , Scavenger Receptors, Class E/metabolism , 3' Untranslated Regions , Apoptosis , Binding Sites , Endothelial Cells/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , MAP Kinase Signaling System/drug effects , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class E/genetics
6.
Can J Physiol Pharmacol ; 92(4): 299-306, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24708212

ABSTRACT

Oxidized low density lipoprotein (oxLDL) induced injury of endothelial cells is considered to be the first step in the pathogenesis of atherosclerosis. This study aimed to investigate some of the effects and mechanisms of puerarin on oxLDL-induced endothelial injuries. We measured cell viability, and the release of lactate dehydrogenase (LDH), nitric oxide (NO), and interleukin-8 (IL-8) to evaluate the protective effects of puerarin. Intracellular reactive oxygen species (ROS) were detected using 2',7'-dichlorofluorescein diacetate (DCFH-DA). The expression of lectin-like low-density lipoprotein receptor-1 (LOX-1), endothelial nitric oxide synthase (eNOS), cyclooxygenase 2 (COX-2), p38MAPK, and protein kinase B (PKB) phosphorylation, nuclear factor-κB (NF-κB) nuclear translocation, and inhibitor of κB (IκB) degradation were detected using quantitative real-time PCR or Western blot. The results showed that oxLDL significantly decreased cell viability, increased LDH and IL-8 release, inhibited NO production, and induced COX-2 expression. Pretreatment with puerarin led to a strong inhibition of these effects. OxLDL stimulated the expression of LOX-1, the overproduction of ROS, the phosphorylation of p38MAPK, the dephosphorylation of PKB, activation of NF-κB, and the degradation of IκB. These oxLDL-induced effects were suppressed after puerarin pretreatment. These results suggest that puerarin inhibits oxLDL-induced endothelial cell injuries, at least in part, via inhibition of the LOX-1-mediated p38MAPK-NF-κB inflammatory and the PKB-eNOS signaling pathways.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Isoflavones/pharmacology , Lipoproteins, LDL/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Scavenger Receptors, Class E/metabolism , Cell Survival/drug effects , Down-Regulation , Enzyme Induction , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipoproteins, LDL/toxicity , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class E/genetics , Signal Transduction , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Pharmazie ; 69(10): 775-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25985569

ABSTRACT

This study was designed to investigate the potential association between NTCP c.800C >T polymorphism and rosuvastatin pharmacokinetics in Chinese healthy males. 305 individuals were enrolled to identify NTCP c.800C > T, OATP1B1 c.521T > C and BCRP c.421C > A genotypes by direct sequencing and pyrosequencing methods, respectively. 17 healthy volunteers who were OATP1B1 c.521TT and BCRP c.421CC wild-type homozygotes with different NTCP c.800C > T genotype were selected to participate in this pharmacokinetic study. Nine were NTCP c.800CC wild-type homozygotes and the other eight subjects were carriers with at least one c.800T variant allele (seven subjects with c.800CT genotype and one was homozygote of c.800TT). All the subjects received a single oral dose of 10 mg rosuvastatin. The plasma concentrations of rosuvastatin were measured up to 72 h by a LC-MS method. NTCP c.800C > T genetic polymorphism markedly effected rosuvastatin pharmacokinetics. The AUC(o-72) and AUC(0 --> ∞) in subjects with NTCP c.800CT + TT genotype were 56% (162.64 ± 37.55 vs. 103.99 ± 28.15 ng x h/ml, P = 0.016) and 57% greater (178.51 ± 42.75 vs. 113.60 ± 33.73 ng x h/ml, P = 0.020) than those in the c.800CC wild-type subjects, respectively. In the c.800CT + TT mutant group, the C(max) was about 78% higher than those in c.800CC genotype (14.31 ± 3.63 vs. 8.04 ± 1.72 ng x h/ml, P = 0.004). The oral clearance (CL/F) of rosuvastatin in subjects with the c.800CT+TT genotype was only 63% of those in the c.800CC genotype (58.32 ± 12.16 vs. 93.04 ± 20.61 ng x h/ml, P = 0.009). The half-time (T1/2) and the T(max) had no significant difference between two groups (p = 0.466 and 0.713, respectively). NTCP c.800C > T polymorphism play a critical role in the individual variability of rosuvastatin pharmacokinetics in Chinese healthy males after excluding the impact of OATP1B1 c.521T > C and BCRP c.421C > A polymorphisms.


Subject(s)
Fluorobenzenes/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Organic Anion Transporters, Sodium-Dependent/genetics , Pyrimidines/pharmacokinetics , Sulfonamides/pharmacokinetics , Symporters/genetics , Adult , Area Under Curve , Asian People , China/epidemiology , Gene Frequency , Humans , Male , Polymorphism, Genetic/genetics , Rosuvastatin Calcium
8.
Int J Mol Sci ; 14(11): 23086-102, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24284400

ABSTRACT

The let-7 family is the second microRNA found in C. elegans. Recent researches have found it is highly expressed in the cardiovascular system. Studies have revealed the aberrant expression of let-7 members in cardiovascular diseases, such as heart hypertrophy, cardiac fibrosis, dilated cardiomyopathy (DCM), myocardial infarction (MI), arrhythmia, angiogenesis, atherosclerosis, and hypertension. Let-7 also participates in cardiovascular differentiation of embryonic stem cells. TLR4, LOX-1, Bcl-xl and AGO1 are by now the identified target genes of let-7. The circulating let-7b is suspected to be the biomarker of acute MI and let-7i, the biomarker of DCM. Further studies are necessary for identifying the gene targets and signaling pathways of let-7 in cardiovascular diseases. Let-7 might be a potential therapeutic target for cardiovascular diseases. This review focuses on the research progresses regarding the roles of let-7 in cardiovascular development and diseases.


Subject(s)
Cardiovascular Diseases/genetics , Embryonic Stem Cells/metabolism , Heart/growth & development , MicroRNAs/genetics , Cardiovascular Diseases/pathology , Cell Differentiation/genetics , Humans , Myocardium/metabolism , Signal Transduction
9.
PLoS One ; 8(10): e76474, 2013.
Article in English | MEDLINE | ID: mdl-24146874

ABSTRACT

Organic cation/carnitine transporter 2 (OCTN2) is responsible for the cellular uptake of the antineoplastic agent, oxaliplatin. Epigenetic modification is a possible mechanism of altered drug-transporter expression in cancers, leading to altered efficacy of chemotherapeutic drugs. However, the mechanisms governing OCTN2 regulation are not completely understood. In this study, the low levels of OCTN2 in HepG2 and LS174T cells were elevated by the demethylating reagent, decitabine (DCA). To further reveal the epigenetic mechanism of down-regulation of OCTN2, we found that Region-1 within the OCTN2 promoter (spanning -354 to +85) was a determinant of OCTN2 expression in a luciferase reporter assay. Moreover, methylation-specific PCR (MSP) and bisulfite genomic sequencing showed that the degree of individual methylated CpG sites within this region was inversely correlated with the levels of OCTN2 in different cancer cells. Application of DCA to HepG2 and LS174T cells reversed the hypermethylation status of the OCTN2 promoter and increased OCTN2 expression, enhancing cellular uptake of oxaliplatin. Thus, we identified that promoter methylation is responsible for epigenetic down-regulation of OCTN2 in HepG2 and LS174T cells. Given the essential role of OCTN2 in cancer cell uptake of chemotherapeutics, and thus treatment efficacy, pretreatment with a demethylating reagent is a possible strategy for optimizing pharmacotherapies against cancers.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Organic Cation Transport Proteins/genetics , Promoter Regions, Genetic , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , COS Cells , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , CpG Islands , Decitabine , Genes, Reporter , Genome, Human/genetics , Humans , Luciferases/metabolism , Molecular Sequence Data , Organic Cation Transport Proteins/metabolism , Organoplatinum Compounds/pharmacology , Oxaliplatin , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solute Carrier Family 22 Member 5 , Transcription, Genetic/drug effects
10.
Asian Pac J Cancer Prev ; 14(1): 387-92, 2013.
Article in English | MEDLINE | ID: mdl-23534758

ABSTRACT

OBJECTIVE: The aim of the present study was to explore mechanisms by which let-7c suppresses NSCLC cell proliferation. METHODS: The expression level of let-7c was quantified by qRT-PCR. A549 and H1299 cells were transfected with let-7c mimics to restore the expression of let-7c. The effects of let-7c were then assessed by cell proliferation, colony formation and cell cycle assay. Mouse experiments were used to confirm the effect of let-7c on tumorigenicity in vivo. Luciferase reporter assays and Western blotting were performed to identify target genes for let-7c. RESULTS: HOXA1 was identified as a novel target of let-7c. MTS, colony formation and flow cytometry assays demonstrated that forced expression of let-7c inhibited NSCLC cell proliferation by inducing G1 arrest in vitro, consistent with inhibitory effects induced by knockdown of HOXA1. Mouse experiments demonstrated that let-7c expression suppressed tumorigenesis. Furthermore, we found that let-7c could regulate the expression of HOXA1 downstream effectors CCND1, CDC25A and CDK2. CONCLUSIONS: Collectively, these results demonstrate let-7c inhibits NSCLC cell proliferation and tumorigenesis by partial direct targeting of the HOXA1 pathway, which suggests that restoration of let-7c expression may thus offer a potential therapeutic intervention strategy for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lung Neoplasms/metabolism , MicroRNAs/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , 3' Untranslated Regions , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival , Cell Transformation, Neoplastic , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Down-Regulation , Flow Cytometry , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation , HEK293 Cells , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , RNA, Messenger/metabolism , Transfection , Tumor Stem Cell Assay , cdc25 Phosphatases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...