Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
FASEB J ; 37(1): e22657, 2023 01.
Article in English | MEDLINE | ID: mdl-36459147

ABSTRACT

Investigations on placental P-glycoprotein (P-gp) regulation could provide more therapeutic targets for individualized and safe pharmacotherapy during pregnancy. The role of long noncoding RNA (lncRNA) on placental P-gp regulation is lacking. The present study was carried out to investigate the regulation and underlying mechanisms of lncRNA urothelial carcinoma associated 1 (UCA1) on P-gp in Bewo cells. lncRNA UCA1 inhibition or overexpression could decrease or increase ABCB1 mRNA expression, P-gp expression and its cellular efflux function, respectively. RNA-FISH revealed that lncRNA UCA1 was mainly located in the cytoplasm of Bewo cells. MicroRNA array was applied and 10 significant miRNAs was identified after lncRNA UCA1 inhibition. Databases of LncTarD, LncRNA2Target, and miRcode were further used to search potential target miRNAs of lncRNA UCA1 and miR-16-5p was screened out. Thereafter, we confirmed that miR-16-5p expression was significantly upregulated or reduced after lncRNA UCA1 knockdown or overexpression, respectively. Furthermore, we also proved that ABCB1 mRNA expression, P-gp expression and its cellular efflux function was enhanced or reduced after miR-16-5p inhibition or overexpression, respectively. The rescue experiment further indicated that miR-16-5p was involved in the positive regulation of lncRNA UCA1 on the expression and function of P-gp. Lastly, dual-luciferase reporter system, RNA-binding protein immunoprecipitation and RNA pull-down assays were performed to explore the relationships among lncRNA UCA1, miR-16-5p, and ABCB1. It was found that lncRNA UCA1(1103-1125) could directly interact with miR-16-5p and miR-16-5p could directly target ABCB1 coding DNA sequence region (882-907). In conclusion, LncRNA UCA1 could promote the expression and function of P-gp by sponging miR-16-5p in BeWo cells.


Subject(s)
Carcinoma, Transitional Cell , MicroRNAs , RNA, Long Noncoding , Urinary Bladder Neoplasms , Pregnancy , Humans , Female , RNA, Long Noncoding/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Placenta , ATP Binding Cassette Transporter, Subfamily B , MicroRNAs/genetics , RNA, Messenger
2.
PLoS One ; 14(5): e0214873, 2019.
Article in English | MEDLINE | ID: mdl-31086358

ABSTRACT

BACKGROUNDS: Reducing toxicants transplacental rates could contribute to the prevention of congenital heart defects (CHDs). Placental P-glycoprotein (P-gp) plays a vital role in fetal toxicants exposure and subsequently affects the risk of toxicants-induced birth defects. However, data on the role of placental P-gp in decreasing toxicants-induced cardiac anomalies is extremely limited. This study aimed to explore the protective role of placental P-gp in reducing the risk of Di-(2-ethylhexyl)-phthalate (DEHP) induced cardiac anomalies in mice. METHODS: The C57BL mice were randomly divided into four groups: the vehicle group (corn oil, n = 10), 500mg/Kg DEHP group (n = 15), 3mg/Kg verapamil group (n = 10) and 500mg/Kg DEHP & 3mg/Kg verapamil group (n = 20). Pregnant dams in different group received respective intervention by gavage once daily from E6.5-14.5. Maternal weights were monitored every day and samples were collected at E15.5. HE staining was used to examine fetal cardiac malformations. Real-time quantitative PCR (RT-qPCR) and Western-Blot were applied to detect Nkx2.5/Gata4/Tbx5/Mef2c/Chf1 mRNA and protein expression, respectively. The mRNA expression of peroxisome proliferator-activated receptor γ (PPARγ) was also determined using RT-qPCR. RESULTS: Co-administration of verapamil and DEHP significantly elevated fetal cardiac malformation rates, in comparison with the DEHP group, the verapamil group and the vehicle group. Different phenotypes of cardiac anomalies, including septal defects and ventricular myocardium noncompaction, were noted both in the DEHP group and the DEHP & verapamil group. The ventricular myocardium noncompaction appeared to be more severe in the DEHP & verapamil group. Fetal cardiac PPARγ mRNA expression was notably increased and Gata4/Mef2c/Chf1 expression was markedly decreased in the DEHP & verapamil group. CONCLUSION: Placental P-gp inhibition enhances susceptibility to DEHP induced cardiac malformations in mice.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Diethylhexyl Phthalate/toxicity , Fetal Heart/drug effects , Heart Defects, Congenital/pathology , Placenta/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Female , Fetal Heart/metabolism , Fetal Heart/pathology , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/drug effects , Heart Defects, Congenital/chemically induced , Heart Defects, Congenital/metabolism , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Maternal Exposure , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/metabolism , Pregnancy , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Verapamil/pharmacology
4.
Medicine (Baltimore) ; 97(21): e10712, 2018 May.
Article in English | MEDLINE | ID: mdl-29794746

ABSTRACT

BACKGROUND: To interpret the various associations between miRNA polymorphisms and cardiovascular diseases (CVD). METHODS: Literature search has identified relevant studies up to June 2016. A meta-analysis was performed followed the guidelines from the Cochrane review group and the PRISMA statement. Studies were identified by searching the Cochrane Library, EMBASE, PUBMED and WHO clinical trials registry center. A meta-analysis has been done with a fixed/random-effect model using STATA 14.0, which also has been used to estimate the publication bias and meta-regression. RESULTS: The results from 11 case-control studies were included. The miR-146a G/C makes a contribution to the causing of CVD as recessive genetic model. And the miR-499 G/A raised the risks of cardiomyopathy, however it could still accelerate the procedure of CVD combined with myocardial infraction. At this point, we consider that it could deepen the adverse of outcomes from coronary artery disease (CAD), but it's hard to draw an association between miR-499 G/A and CAD. At last the miR-196a2 T/C demonstrated a contrary role between development problem and metabolic issues, which protects the development procedure and impairs the metabolism to cause different disease phenotypes. CONCLUSION: Despite inter-study variability, the polymorphisms from miR-146a, miR-499 and miR-196a2 have impacts on cardiovascular disease. Each type of miRNA has individual role in either cardiac development or the origins of CVD.


Subject(s)
Cardiovascular Diseases/genetics , MicroRNAs/genetics , Polymorphism, Genetic/genetics , Genetic Predisposition to Disease , Humans , Risk Factors
5.
J Appl Toxicol ; 38(6): 834-842, 2018 06.
Article in English | MEDLINE | ID: mdl-29377175

ABSTRACT

Accumulating evidence has suggested a link between maternal di-(2-ethylhexyl)-phthalate (DEHP) exposure and various developmental abnormalities. However, the evidence regarding the effect of maternal DEHP exposure on fetal cardiac development is scarce. The present study aimed to determine the effect of maternal DEHP exposure on fetal cardiac development in mice and explore the possible involved mechanism preliminarily. The C57BL mice were randomly divided into four groups: the vehicle group (corn oil, n = 10), 250 mg kg-1 DEHP group (n = 15), 500 mg kg-1 DEHP group (n = 20) and 1 g kg-1 DEHP group (n = 20). Pregnant dams in different group received respective intervention by gavage once daily from embryonic day (E)6.5 to E14.5. Maternal weights were monitored every day and samples were collected at E15.5. Hematoxylin and eosin staining was used to examine fetal cardiac malformations. Real-time quantitative polymerase chain reaction and western blot were applied to detect peroxisome proliferator-activated receptor (PPAR)α/PPARγ/Nkx2.5/Gata4/Tbx5/Mef2c/Chf1 mRNA and protein expression, respectively. Maternal DEHP exposure significantly decreased maternal body weight, fetal weight and placental weight, and remarkably elevated fetal cardiac malformations rate. The phenotypes of cardiac anomalies mainly include septal defects, ventricular myocardium noncompaction and cardiac hypoplasia. Higher doses DEHP (500 mg kg-1 and 1 g kg-1 ) could significantly decreased fetal cardiac Gata4/Mef2c/Chf1 expression, while PPARγ expression was upregulated. Maternal exposure to higher doses of DEHP could result in fetal cardiac development malformations in mice and it might have resulted from the inhibition of cardiac GATA4/Mef2c/Chf1 expression via PPARγ activation.


Subject(s)
Diethylhexyl Phthalate/toxicity , Fetal Heart/drug effects , Heart Defects, Congenital/chemically induced , Plasticizers/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Fetal Heart/growth & development , Fetal Heart/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/physiopathology , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Maternal Exposure , Mice, Inbred C57BL , Morphogenesis/drug effects , PPAR gamma/agonists , PPAR gamma/genetics , PPAR gamma/metabolism , Pregnancy , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/drug effects
6.
Placenta ; 58: 105-114, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28962688

ABSTRACT

INTRODUCTION: Placental P-glycoprotein (P-gp) plays a significant role in regulating drugs' transplacental transfer rates. Investigations on placental P-gp regulation could provide more therapeutic targets for individualized and safe pharmacotherapy during pregnancy. Currently, the epigenetic regulation of placental P-gp is rare. Our previous study has demonstrated that HDACs inhibition could up-regulate placental P-gp and HDAC1/2/3 might be involved in this process. The present study was carried out to further explore whether HDAC1/2/3 were indeed involved in the regulation of placental P-gp or not and screen out the subtype engaged in this process. METHODS: BeWo and JAR cells were transfected with HDAC1/2/3 specific siRNA. After 48 h of transfection, cells were harvested for real-time quantitative PCR (qRT-PCR), Western blot, immunofluorescence and fluorescent dye efflux assay to evaluate P-gp expression, localization, and efflux activity, respectively. Hdac2 siRNA was intraperitoneally injected to pregnant mice every 48 h from E7.5 to E15.5 and digoxin was administered by gavages 1 h prior to euthanasia at E16.5. Placental Hdac1/2/3 and P-gp expression were determined by qRT-PCR and Western blot. Maternal plasma and fetal-unit digoxin concentrations were detected by enzyme-multiplied immunoassay. RESULTS: In vitro, HDAC2 inhibition could significantly elevate P-gp expression and reduce intracellular accumulation of P-gp substrates (DiOC2 (3) and Rh 123) both in BeWo and JAR, while knockdown of HDAC1/3 had no influence on P-gp expression and its efflux activity. Additionally, in vivo, Hdac2 silencing in pregnant mice also elevated placental P-gp expression and decreased digoxin transplacental transfer rate. CONCLUSION: HDAC2 inhibition could result in induction of placental P-gp expression and functionality both in vitro and in vivo.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Histone Deacetylase 2/metabolism , Placenta/metabolism , Animals , Cell Line, Tumor , Digoxin/pharmacokinetics , Epigenesis, Genetic , Female , Histone Deacetylase 2/genetics , Humans , Mice , Placenta/drug effects , Pregnancy , RNA, Small Interfering
7.
Medicine (Baltimore) ; 96(22): e6825, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28562533

ABSTRACT

BACKGROUND: With the rapid development of molecular biology, the kind of mircoRNA (miRNA) has been introduced into emerging role both in cardiac development and pathological procedure. Thus, we conduct this meta-analysis to find out the role of circulating miRNA as a biomarker in detecting heart failure. METHODS: We searched PubMed, EMBASE, the Cochrane Central Register of Controlled Trials, and World Health Organization clinical trials registry center to identify relevant studies up to August 2016. We performed meta-analysis in a fixed/random-effect model using Meta-disc 1.4. We used STATA 14.0 to estimate the publication bias and meta-regression. Besides, we took use of SPSS 17.0 to evaluate variance between several groups. Information on true positive, false positive, false negative, and true negative, as well as the quality of research was extracted. RESULTS: We use results from 10 articles to analyze the pooled accuracy. The overall performance of total mixed miRNAs (TmiRs) detection was: pooled sensitivity, 0.74 (95% confidence interval [CI], 0.72 to 0.75); pooled specificity, 0.69 (95%CI, 0.67 to 0.71); and area under the summary receiver operating characteristic curves value (SROC), 0.7991. The miRNA-423-5p (miR-423-5p) detection was: pooled sensitivity, 0.81 (95%CI, 0.76 to 0.85); pooled specificity, 0.67 (95%CI, 0.61 to 0.73); and SROC, 0.8600. However, taken the same patients population, we extracted the data of BNP for detecting heart failure and performed meta-analysis with acceptable SROC as 0.9291. Among the variance analysis, the diagnostic performance of miR-423-5p claimed significant advantages of other pooled results. However, the combination of miRNAs and BNP could increase the accuracy of detecting of heart failure. Unfortunately, there was no dramatic advantage of miR-423-5p compared to BNP protocol. CONCLUSION: Despite interstudy variability, the performance test of miRNA for detecting heart failure revealed that miR-423-5p demonstrated the potential to be a biomarker. However, other miRNAs were not able to provide enough evidence on promising diagnostic value for heart failure based on the current data. Moreover, the combination of miRNAs and BNP could work as a better method to detection. Unfortunately, BNP was still the most convinced biomarker for such disease.


Subject(s)
Heart Failure/blood , MicroRNAs/blood , Biomarkers/blood , Humans
8.
Zhongguo Dang Dai Er Ke Za Zhi ; 19(4): 475-483, 2017 Apr.
Article in Chinese | MEDLINE | ID: mdl-28407839

ABSTRACT

OBJECTIVE: To investigate the effect of histone acetylation/deacetylation imbalances on embryonic hearts of mice and its effect on key genes of planar cell polarity (PCP) pathway-Vangl2, Scrib and Rac1 in H9C2 cells. METHODS: Forty pregnant C57/B6 mice were randomly assigned into three groups: blank group (n=10), vehicle group (n=10), and valproic acid (VPA)-treated group (n=20). In the VPA-treated group, VPA, a histone deacetylase (HDAC) inhibitor, was administered to each individual dam intraperitoneally at a single dose of 700 mg/kg on embryonic day 10.5 (E10.5). The vehicle and blank groups received equivalent saline or no interventions, respectively. Dams were sacrificed on E15.5, and death rates of embryos were evaluated. Subsequently, embryonic hearts of survival fetus were removed to observe cardiac abnormalities by hematoxylin-eosin (HE) staining. H9C2 cells were cultured and allotted to the blank, vehicle, and VPA-treated groups: the VPA treated group received VPA exposure at concentrations of 2.0, 4.0 and 8.0 mmol/L; the vehicle and blank groups received equivalent saline or no interventions, respectively. HDAC1-3 as well as Vangl2, Scrib and Rac1 mRNA and protein expression levels were determined by quantitative real-time PCR and Western blot, respectively. The total HDAC activity was analyzed by colorimetric assay. RESULTS: The fetus mortality rate after VPA treatment was 31.7%, with a significantly higher rate of cardiac abnormalities in comparison with the controls (P<0.05). In comparison with the blank and vehicle groups, HDAC1 mRNA was significantly increased at various concentrations of VPA treatment at all time points of exposure (P<0.05), together with a reduction of protein level after 48 and 72 hours of exposure (P<0.05). The inhibition of HDAC2 mRNA after various concentrations of VPA incubation was pronounced at 24 hours of exposure (P<0.05), while the protein levels were reduced at all time points (P<0.05). HDAC3 mRNA was prominently induced by VPA (4.0 and 8.0 mmol/L) at all time points of treatment (P<0.05). In contrast, the protein level was inhibited after VPA treatment (P<0.05). In comparison with the blank and vehicle groups, Vangl2 mRNA as well as Scrib mRNA/protein expression levels were markedly reduced after 48 and 72 hours of VPA treatment (P<0.05), together with a reduction of protein level in Vangl2 at 72 hours (P<0.05). Compared with the blank and vehicle groups, a significant repression in the total HDAC activity was observed in the VPA-treated group at concentrations of 4.0 and 8.0 mmol/L after 24 hours of treatment (P<0.05), and the effect persisted up to 48 and 72 hours, exhibiting pronounced inhibition at all concentrations (P<0.05). CONCLUSIONS: VPA might result in acetylation/deacetylation imbalances by inhibiting HDAC1-3 protein expression and total HDAC activity, leading to the down-regulation of mRNA and protein expression of Vangl2 and Scrib. This could be one of the mechanisms contributing to congenital heart disease.


Subject(s)
Fetal Heart/metabolism , Histones/metabolism , Acetylation , Animals , Cell Polarity , Cells, Cultured , Fetal Heart/drug effects , Heart Defects, Congenital/etiology , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , RNA, Messenger/analysis , Valproic Acid/pharmacology
9.
Placenta ; 49: 37-47, 2017 01.
Article in English | MEDLINE | ID: mdl-28012453

ABSTRACT

INTRODUCTIONS: Placental P-glycoprotein (P-gp), encoded by ABCB1 gene in human, plays a significant role in regulating drugs' transplacental transfer rates. Investigations on placental P-gp regulation could provide more therapeutic targets for individualized and safe pharmacotherapy during pregnancy. Currently, the epigenetic control of placental P-gp is rare. This study aimed to investigate the effect of histone deacetylases (HDACs) inhibition on P-gp expression in placental trophoblast cell lines and to explore whether HDAC1/2/3 was involved in this process preliminarily. METHODS: Human placental trophoblast cell lines (Bewo and JAR) were treated with two different HDAC inhibitors-suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA) at different concentration gradients of 0.5, 1.0, 3.0 and 5.0 µM. Cells were harvested after 24, 48, and 72 h treatment. Total HDAC activity was detected by colorimetric assay Kits. HDAC1/2/3/ABCB1 mRNA and protein expressions were determined by real-time quantitative PCR and western-blot, respectively. Pearson correlation analysis test was performed to explore the relationship between HDAC1/2/3 mRNA and ABCB1 mRNA expression. RESULTS: SAHA and TSA could inhibit total HDAC activity and placental HDAC1/2/3 expression both in Bewo and JAR, but displayed a transient induction of HDAC mRNA or protein level after being treated at low dosage or prolonged exposure to drugs. Discordance in HDAC mRNA and protein expression was also observed. Placental P-gp expression was significantly induced in company with HDACs inhibition. There was a significant negative linear relationship between HDAC1/2 mRNA and ABCB1 mRNA expression. CONCLUSIONS: HDACs inhibition could up-regulate placental P-gp expression in trophoblast cells, and HDAC1/2 was most likely to be involved in this process.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Trophoblasts/drug effects , Cell Line , Female , Humans , Hydroxamic Acids/pharmacology , Pregnancy , Trophoblasts/cytology , Trophoblasts/metabolism , Vorinostat
10.
Placenta ; 44: 91-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27452443

ABSTRACT

INTRODUCTION: Placental P-glycoprotein (P-gp) plays a significant role in controlling transplacental digoxin transfer rate. Investigations on P-gp regulation in placenta of women with different pregnant pathological states are of great significance to individualized transplacental digoxin treatment for fetal heart failure (FHF). This study aimed to explore the effect of 17α-ethynylestradiol induced intrahepatic cholestasis of pregnancy (ICP) on placental P-gp in mice. METHODS: ICP model in mice was induced by subcutaneous injection of 17α-ethynylestradiol dissolved in propylene glycol once daily from E12.5 to E16.5. Maternal plasma ALT, AST, TB, DBIL, γ-GT, LDH, ALP and TBA concentrations were measured. HE staining was applied for observation of maternal liver cells degeneration, necrosis and intrahepatic cholestasis. Placental Abcb1a/Abcb1b/HIF-1α mRNA and P-gp/HIF-1α protein expression were determined by real-time quantitative PCR and western-blot. Maternal plasma and fetal-unit digoxin concentrations were detected by a commercial kit assay. RESULTS: The ICP group showed higher levels of maternal plasma ALT, AST, TB, DBIL, γ-GT, LDH, ALP and TBA concentrations, reduction in fetal survival rates, lower placental and fetal weights, and typical liver cells degeneration, necrosis and intrahepatic cholestasis. The placental Abcb1a mRNA and P-gp expression of ICP group were significantly elevated, while transplacental digoxin transfer rates were significantly decreased. Both placental HIF-1α mRNA and protein expression was significantly elevated in the ICP group, and there was a positive correlation between Abcb1a mRNA and HIF-1α mRNA. CONCLUSIONS: 17α-ethynylestradiol induced ICP could up-regulate placental P-gp expression and reduce transplacental digoxin transfer rate in mice, which might be partly associated with higher expression of HIF-1α.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cholestasis, Intrahepatic/metabolism , Digoxin/therapeutic use , Heart Failure/metabolism , Placenta/metabolism , Pregnancy Complications/metabolism , Animals , Cholestasis, Intrahepatic/chemically induced , Cholestasis, Intrahepatic/pathology , Ethinyl Estradiol , Female , Heart Failure/drug therapy , Heart Failure/pathology , Mice , Placenta/pathology , Pregnancy , Pregnancy Complications/chemically induced , Pregnancy Complications/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...