14 results on '"Cindy Yanfei Li"'
Search Results
2. Contribution of Uptake and Efflux Transporters to Oral Pharmacokinetics of Furosemide
- Author
-
Mayur K. Ladumor, Bhagwat Prasad, Aarzoo Thakur, Revathi Chapa, Sheena Sharma, Arzu Selen, Abdul Basit, Cindy Yanfei Li, and Saranjit Singh
- Subjects
Physiologically based pharmacokinetic modelling ,Organic anion transporter 1 ,biology ,Chemistry ,General Chemical Engineering ,Multidrug resistance-associated protein 2 ,medicine.medical_treatment ,Furosemide ,General Chemistry ,Pharmacology ,Article ,Bioavailability ,Organic anion-transporting polypeptide ,Pharmacokinetics ,biology.protein ,medicine ,Diuretic ,QD1-999 ,medicine.drug - Abstract
Furosemide is a widely used diuretic for treating excessive fluid accumulation caused by disease conditions like heart failure and liver cirrhosis. Furosemide tablet formulation exhibits variable pharmacokinetics (PK) with bioavailability ranging from 10 to almost 100%. To explain the variable absorption, we integrated the physicochemical, in vitro dissolution, permeability, distribution, and the elimination parameters of furosemide in a physiologically-based pharmacokinetic (PBPK) model. Although the intravenous PBPK model reasonably described the observed in vivo PK data, the reported low passive permeability failed to capture the observed data after oral administration. To mechanistically justify this discrepancy, we hypothesized that transporter-mediated uptake contributes to the oral absorption of furosemide in conjunction with passive permeability. Our in vitro results confirmed that furosemide is a substrate of intestinal breast cancer resistance protein (BCRP), multidrug resistance-associated protein 4 (MRP4), and organic anion transporting polypeptide 2B1 (OATP2B1), but it is not a substrate of P-glycoprotein (P-gp) and MRP2. We then estimated the net transporter-mediated intestinal uptake and integrated it into the PBPK model under both fasting and fed conditions. Our in vitro data and PBPK model suggest that the absorption of furosemide is permeability-limited, and OATP2B1 and MRP4 are important for its permeability across intestinal membrane. Further, as furosemide has been proposed as a probe substrate of renal organic anion transporters (OATs) for assessing clinical drug-drug interactions (DDIs) during drug development, the confounding effects of intestinal transporters identified in this study on furosemide PK should be considered in the clinical transporter DDI studies.
- Published
- 2020
3. Organic Anion Transporting Polypeptide–Mediated Hepatic Uptake of Glucuronide Metabolites of Androgens
- Author
-
Zsuzsanna Gáborik, Emese Kis, Bhagwat Prasad, Anshul Gupta, and Cindy Yanfei Li
- Subjects
Proteomics ,0301 basic medicine ,Androsterone glucuronide ,medicine.drug_class ,Organic Anion Transporters ,Cell Line ,Solute Carrier Organic Anion Transporter Family Member 1B3 ,03 medical and health sciences ,chemistry.chemical_compound ,Glucuronides ,0302 clinical medicine ,medicine ,Humans ,Testosterone glucuronide ,Pharmacology ,Etiocholanolone ,Androsterone ,biology ,Liver-Specific Organic Anion Transporter 1 ,Chemistry ,Biological Transport ,Androgen ,Organic anion-transporting polypeptide ,HEK293 Cells ,030104 developmental biology ,Liver ,Biochemistry ,Dihydrotestosterone ,Androgens ,biology.protein ,Molecular Medicine ,Glucuronide ,030217 neurology & neurosurgery ,medicine.drug - Abstract
We previously established that androgen glucuronides are primarily effluxed by MRP2 and MRP3 in liver and intestine. However, no data exist on the mechanism of hepatic uptake of these metabolites. To fill this knowledge gap, the first goal of this study was to explore the role of hepatic uptake transporters and characterize transport kinetics of glucuronides of testosterone (TG), dihydrotestosterone (DHTG), androsterone (AG), and etiocholanolone (EtioG) using cell-lines overexpressing organic anion transporting polypeptide (OATP1B1, OATP1B3, and OATP2B1). Using quantitative proteomics-guided approach, we then estimated the relative contribution (ft) of individual OATPs in hepatic uptake of the androgen glucuronides. The transport screening assays revealed that the glucuronides were primarily influxed by OATP1B1 and OATP1B3. The Km values for OATP1B1-mediated uptake were low for EtioG (6.2 µM), followed by AG, TG, and DHTG (46.2, 56.7, and 71.3 µM, respectively). Whereas, the Km value for OATP1B3-mediated uptake for EtioG, AG, DHTG, and TG were 19.8, 29.3, 69.6, and 110.4 µM, respectively. Both OATP1B1 and OATP1B3 exhibited highest transport rate toward AG as compared to other glucuronides. When adjusted for transporter abundance in human livers, EtioG and DHTG were transported equally by OATP1B1 and OATP1B3, whereas TG and AG were preferentially (>68%) transported by OATP1B3. Collectively, this is the first report that elucidate mechanisms of hepatic uptake of androgen glucuronides. Perturbation in these processes by genetic polymorphisms, disease conditions or drug-interactions can lead to changes in enterohepatic recycling of androgen. The apparent higher selectivity of OATP1B3 towards TG and AG can be leveraged for establishing these metabolites as clinical biomarkers of OATP1B3 activity. SIGNIFICANCE STATEMENT This is the first study to elucidate the mechanism of hepatic uptake of androgen glucuronides and estimate the relative contribution (ft) of individual OATPs using quantitative proteomics. Our results show that both OATP1B1 and OATP1B3 are responsible for the hepatic uptake of major circulating testosterone glucuronides. The apparent higher selectivity of OATP1B3 towards testosterone glucuronide and androsterone glucuronide can be leveraged for establishing these metabolites as clinical biomarkers of OATP1B3 activity.
- Published
- 2020
- Full Text
- View/download PDF
4. Regulation of Drug Transport Proteins-From Mechanisms to Clinical Impact: A White Paper on Behalf of the International Transporter Consortium
- Author
-
Kim L.R. Brouwer, Raymond Evers, Elizabeth Hayden, Shuiying Hu, Cindy Yanfei Li, Henriette E. Meyer zu Schwabedissen, Sibylle Neuhoff, Stefan Oswald, Micheline Piquette‐Miller, Chitra Saran, Noora Sjöstedt, Jason A. Sprowl, Simone H. Stahl, and Wei Yue
- Subjects
Pharmacology ,Gene Expression Regulation ,Pharmaceutical Preparations ,Humans ,Membrane Transport Proteins ,Receptors, Cytoplasmic and Nuclear ,Pharmacology (medical) ,Biological Transport ,Carrier Proteins - Abstract
Membrane transport proteins are involved in the absorption, disposition, efficacy, and/or toxicity of many drugs. Numerous mechanisms (e.g., nuclear receptors, epigenetic gene regulation, microRNAs, alternative splicing, post-translational modifications, and trafficking) regulate transport protein levels, localization, and function. Various factors associated with disease, medications, and dietary constituents, for example, may alter the regulation and activity of transport proteins in the intestine, liver, kidneys, brain, lungs, placenta, and other important sites, such as tumor tissue. This white paper reviews key mechanisms and regulatory factors that alter the function of clinically relevant transport proteins involved in drug disposition. Current considerations with in vitro and in vivo models that are used to investigate transporter regulation are discussed, including strengths, limitations, and the inherent challenges in predicting the impact of changes due to regulation of one transporter on compensatory pathways and overall drug disposition. In addition, translation and scaling of in vitro observations to in vivo outcomes are considered. The importance of incorporating altered transporter regulation in modeling and simulation approaches to predict the clinical impact on drug disposition is also discussed. Regulation of transporters is highly complex and, therefore, identification of knowledge gaps will aid in directing future research to expand our understanding of clinically relevant molecular mechanisms of transporter regulation. This information is critical to the development of tools and approaches to improve therapeutic outcomes by predicting more accurately the impact of regulation-mediated changes in transporter function on drug disposition and response.
- Published
- 2022
5. Prediction of Hepatobiliary Clearances and Hepatic Concentrations of Transported Drugs in Humans Using Rosuvastatin as a Model Drug
- Author
-
Flavia Storelli, Cindy Yanfei Li, Madhav Sachar, Vineet Kumar, Scott Heyward, Zsolt Sáfár, Emese Kis, and Jashvant D. Unadkat
- Subjects
Pharmacology ,Liver ,Hepatocytes ,ATP Binding Cassette Transporter, Subfamily G, Member 2 ,Humans ,Pharmacology (medical) ,Biological Transport ,Rosuvastatin Calcium ,Neoplasm Proteins - Abstract
To assess efficacy and toxicity of a drug in humans, it is important to measure the tissue concentration of a drug at the target site. For a drug that is transported into or out of the tissue, the tissue unbound steady-state concentration can be dramatically different from its corresponding unbound steady-state plasma concentration. Because routine measurement of drug tissue concentrations is not possible, using rosuvastatin as a model transporter substrate drug, we compared the ability of the proteomics-informed relative expression factor (REF) approach and sandwich-cultured human hepatocytes (SCH) to accurately predict rosuvastatin human hepatobiliary clearances and hepatic concentrations. REF-predicted rosuvastatin biliary clearance (CL
- Published
- 2021
6. Polybrominated Diphenyl Ethers and Gut Microbiome Modulate Metabolic Syndrome–Related Aqueous Metabolites in Mice
- Author
-
Cindy Yanfei Li, Daniel Raftery, Sridhar Mani, Haiwei Gu, David K. Scoville, Joseph L. Dempsey, Julia Yue Cui, and Dongfang Wang
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Glycosylation ,Administration, Oral ,Pharmaceutical Science ,Mannose ,Hydroxylation ,Mannosyltransferases ,030226 pharmacology & pharmacy ,Transcriptome ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Polybrominated diphenyl ethers ,Internal medicine ,Halogenated Diphenyl Ethers ,medicine ,Animals ,Germ-Free Life ,Humans ,Intestine, Large ,Microbiome ,reproductive and urinary physiology ,Metabolic Syndrome ,Pharmacology ,Articles ,medicine.disease ,Glycome ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,Disease Models, Animal ,Tryptophan Metabolite ,Endocrinology ,Liver ,chemistry ,030220 oncology & carcinogenesis ,Dysbiosis ,Environmental Pollutants ,Metabolic syndrome - Abstract
Polybrominated diphenyl ethers (PBDEs) are persistent environmental toxicants associated with increased risk for metabolic syndrome. Intermediary metabolism is influenced by the intestinal microbiome. To test the hypothesis that PBDEs reduce host-beneficial intermediary metabolites in an intestinal microbiome–dependent manner, 9-week old male conventional (CV) and germ-free (GF) C57BL/6 mice were orally gavaged once daily with vehicle, BDE-47, or BDE-99 (100 μmol/kg) for 4 days. Intestinal microbiome (16S rDNA sequencing), liver transcriptome (RNA-Seq), and intermediary metabolites in serum, liver, as well as small and large intestinal contents (SIC and LIC; LC-MS) were examined. Changes in intermediary metabolite abundances in serum, liver, and SIC, were observed under basal conditions (CV vs. GF mice) and by PBDE exposure. PBDEs altered the largest number of metabolites in the LIC; most were regulated by PBDEs in GF conditions. Importantly, intestinal microbiome was necessary for PBDE-mediated decreases in branched-chain and aromatic amino acid metabolites, including 3-indolepropionic acid, a tryptophan metabolite recently shown to be protective against inflammation and diabetes. Gene-metabolite networks revealed a positive association between the hepatic glycan synthesis gene α-1,6-mannosyltransferase (Alg12) mRNA and mannose, which are important for protein glycosylation. Glycome changes have been observed in patients with metabolic syndrome. In LIC of CV mice, 23 bacterial taxa were regulated by PBDEs. Correlations of certain taxa with distinct serum metabolites further highlight a modulatory role of the microbiome in mediating PBDE effects. In summary, PBDEs impact intermediary metabolism in an intestinal microbiome–dependent manner, suggesting that dysbiosis may contribute to PBDE-mediated toxicities that include metabolic syndrome.
- Published
- 2019
- Full Text
- View/download PDF
7. Pitfalls in Predicting Hepatobiliary Drug Transport Using Human Sandwich-Cultured Hepatocytes
- Author
-
Zsuzsanna Gáborik, Kazuya Ishida, Emese Kis, Cindy Yanfei Li, Vineet Kumar, and Jashvant D. Unadkat
- Subjects
Drug ,media_common.quotation_subject ,Cell ,Cell Culture Techniques ,Pharmaceutical Science ,Pharmacology ,030226 pharmacology & pharmacy ,Tight Junctions ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,medicine ,Humans ,Rosuvastatin ,Pharmacokinetics ,media_common ,Drug transport ,Tight junction ,Transporter ,Taurocholic acid ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Hepatocytes ,Calcium ,medicine.drug - Abstract
During drug development, in vivo human biliary drug clearances (CL) are usually predicted using human sandwich-cultured hepatocytes (SCH). To do so, SCH are pre-incubated with Ca2+-containing or Ca2+-free buffer to maintain or disrupt canalicular tight junctions (CTJ), respectively. Drug uptake into SCH is then conducted in the presence of Ca2+ (up to 20 min). Under this standard protocol, two key assumptions are made: first, that the CTJ are not reformed during the uptake phase when Ca2+ is repleted, and second, disruption of CTJ by the Ca2+-free buffer does not affect the activity of any of the transporters present in the sinusoidal or canalicular membrane. Here we investigated the validity of these assumptions using rosuvastatin (RSV) and taurocholic acid (TCA) as our model drugs. In human SCH, the disrupted CTJ were “reformed” with just 10-min Ca2+ repletion as reflected in a significant increase in TCA cell accumulation. To avoid CTJ reformation and cell toxicity, the standard SCH protocol was modified by conducting the uptake in the absence of Ca2+ for 10 min. Surprisingly, using this protocol, RSV uptake into SCH, plated hepatocytes, and transporter-expressing cells confirmed that Ca2+ depletion substantially decreased NTCP and not OATP1B1 activity. Collectively, this study provides the first evidence of reformation of CTJ in human SCH with 20-min Ca2+ repletion, whereas Ca2+ depletion, during the uptake phase, leads to a significant reduction in NTCP uptake. Thus, the entire SCH protocol needs to be re-examined and optimized to correctly estimate hepatobiliary CL of drugs including those that are NTCP substrates.
- Published
- 2020
8. Novel Interactions between Gut Microbiome and Host Drug-Processing Genes Modify the Hepatic Metabolism of the Environmental Chemicals Polybrominated Diphenyl Ethers
- Author
-
Bhagwat Prasad, Irvin R. Schultz, Sara Cade, Julia Yue Cui, Deepak Kumar Bhatt, Cindy Yanfei Li, Soowan Lee, Theo K. Bammler, and Li-Jung Kuo
- Subjects
Male ,0301 basic medicine ,endocrine system ,CYP3A ,Polybrominated Biphenyls ,Down-Regulation ,Pharmaceutical Science ,Biology ,Hydroxylation ,Microbiology ,Mice ,03 medical and health sciences ,Metabolomics ,Polybrominated diphenyl ethers ,Cytochrome P-450 Enzyme System ,Downregulation and upregulation ,Halogenated Diphenyl Ethers ,Animals ,Biotransformation ,Pharmacology ,Sequence Analysis, RNA ,CYP1A2 ,Articles ,Enzyme assay ,Gastrointestinal Microbiome ,Specific Pathogen-Free Organisms ,Up-Regulation ,Mice, Inbred C57BL ,030104 developmental biology ,Liver ,Biochemistry ,biology.protein ,Environmental Pollutants ,Enterotype ,Drug metabolism - Abstract
The gut microbiome is a novel frontier in xenobiotic metabolism. Polybrominated diphenyl ethers (PBDEs), especially BDE-47 (2, 2′, 4, 4′-tetrabromodiphenyl ether) and BDE-99 (2, 2′, 4, 4′,5-pentabromodiphenyl ether), are among the most abundant and persistent environmental contaminants that produce a variety of toxicities. Little is known about how the gut microbiome affects the hepatic metabolism of PBDEs and the PBDE-mediated regulation of drug-processing genes (DPGs) in vivo. The goal of this study was to determine the role of gut microbiome in modulating the hepatic biotransformation of PBDEs. Nine-week-old male C57BL/6J conventional (CV) or germ-free (GF) mice were treated with vehicle, BDE-47 or BDE-99 (100 μmol/kg) for 4 days. Following BDE-47 treatment, GF mice had higher levels of 5-OH-BDE-47 but lower levels of four other metabolites in liver than CV mice; whereas following BDE-99 treatment GF mice had lower levels of four minor metabolites in liver than CV mice. RNA sequencing demonstrated that the hepatic expression of DPGs was regulated by both PBDEs and enterotypes. Under basal conditions, the lack of gut microbiome upregulated the Cyp2c subfamily but downregulated the Cyp3a subfamily. Following PBDE exposure, certain DPGs were differentially regulated by PBDEs in a gut microbiome–dependent manner. Interestingly, the lack of gut microbiome augmented PBDE-mediated upregulation of many DPGs, such as Cyp1a2 and Cyp3a11 in mouse liver, which was further confirmed by targeted metabolomics. The lack of gut microbiome also augmented the Cyp3a enzyme activity in liver. In conclusion, our study has unveiled a novel interaction between gut microbiome and the hepatic biotransformation of PBDEs.
- Published
- 2017
- Full Text
- View/download PDF
9. Transcriptomic profiling of PBDE-exposed HepaRG cells unveils critical lncRNA- PCG pairs involved in intermediary metabolism
- Author
-
Edward J. Kelly, Lianne Sheppard, Angela Zhang, Julia Yue Cui, and Cindy Yanfei Li
- Subjects
0301 basic medicine ,Cell signaling ,Physiology ,Receptors, Cytoplasmic and Nuclear ,Gene Expression ,Signal transduction ,Pathology and Laboratory Medicine ,Biochemistry ,Transcriptome ,0302 clinical medicine ,Drug Metabolism ,Transcription (biology) ,Gene expression ,Transcriptional regulation ,Halogenated Diphenyl Ethers ,Medicine and Health Sciences ,Bile ,Immune Response ,Flame Retardants ,Regulation of gene expression ,0303 health sciences ,Pregnane X receptor ,Multidisciplinary ,Pregnane X Receptor ,Glutathione ,Cell biology ,Body Fluids ,Nucleic acids ,STAT signaling ,030220 oncology & carcinogenesis ,Sirtuin ,Medicine ,Carbohydrate Metabolism ,RNA, Long Noncoding ,Anatomy ,Research Article ,Science ,Immunology ,Biology ,Cell Line ,03 medical and health sciences ,Signs and Symptoms ,Diagnostic Medicine ,Genetics ,Humans ,PPAR alpha ,Gene Regulation ,Pharmacokinetics ,Non-coding RNA ,Gene ,Constitutive Androstane Receptor ,030304 developmental biology ,Pharmacology ,Inflammation ,Retinoid X Receptor alpha ,Biology and life sciences ,Gene Expression Profiling ,Lipid metabolism ,Lipid Metabolism ,Introns ,030104 developmental biology ,Gene Expression Regulation ,13. Climate action ,biology.protein ,Hepatocytes ,Long non-coding RNAs ,RNA ,Peptides - Abstract
Polybrominated diphenyl ethers (PBDEs) were formally used as flame-retardants and are chemically stable, lipophlic persistent organic pollutants which are known to bioaccumulate in humans. Although its toxicities are well characterized, little is known about the changes in transcriptional regulation caused by PBDE exposure. Long non-coding RNAs (lncRNAs) are increasingly recognized as key regulators of transcriptional and translational processes. It is hypothesized that lncRNAs can regulate nearby protein-coding genes (PCGs) and changes in the transcription of lncRNAs may act incisto perturb gene expression of its neighboring PCGs. The goals of this study were to 1) characterize PCGs and lncRNAs that are differentially regulated from exposure to PBDEs; 2) identify PCG-lncRNA pairs through genome annotation and predictive binding tools; and 3) determine enriched canonical pathways caused by differentially expressed lncRNA-PCGs pairs. HepaRG cells, which are human-derived hepatic cells that accurately represent gene expression profiles of human liver tissue, were exposed to BDE-47 and BDE-99 at a dose of 25 μM for 24 hours. Differentially expressed lncRNA-PCG pairs were identified through DESeq2 and HOMER; significant canonical pathways were determined through Ingenuity Pathway Analysis (IPA). LncTar was used to predict the binding of 19 lncRNA-PCG pairs with known roles in drug-processing pathways. Genome annotation revealed that the majority of the differentially expressed lncRNAs map to PCG introns. PBDEs regulated overlapping pathways with PXR and CAR such as protein ubiqutination pathway and PPARα-RXRα activation but also regulate distinctive pathways involved in intermediary metabolism. BDE-47 uniquely regulated signaling by Rho Family GTPases and PBDE-99 uniquely regulates JAK/Stat signaling, bile acid biosynthesis, sirtuin signaling pathway, and autophagy. In conclusion, lncRNAs play essential roles in modifying important pathways involved in intermediary metabolism such as carbohydrate and lipid metabolism.
- Published
- 2019
10. A11 - Optimized renal transporter quantification by using Aquaporin 1 and Aquaporin 2 as anatomical markers: Application in characterizing the ontogeny of renal transporters and its correlation with hepatic transporters in paired sample
- Author
-
J. Steven Leeder, Cindy Yanfei Li, Bhagwat Prasad, Jashvant D. Unadkat, Abdul Basit, and Chelsea Hosey-Cojocari
- Subjects
Pharmacology ,Paired samples ,Aquaporin 2 ,Ontogeny ,Aquaporin 1 ,Pharmaceutical Science ,Pharmacology (medical) ,Transporter ,Biology ,Renal transporters ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
11. PBDEs Altered Gut Microbiome and Bile Acid Homeostasis in Male C57BL/6 Mice
- Author
-
Kris M. Weigel, Qiang Fei, Bhagwat Prasad, Daniel Raftery, Soo Wan Lee, Haiwei Gu, Julia Yue Cui, Dongfang Wang, Deepak Kumar Bhatt, Cindy Yanfei Li, and Joseph L. Dempsey
- Subjects
0301 basic medicine ,Male ,medicine.drug_class ,Pharmaceutical Science ,Down-Regulation ,010501 environmental sciences ,Biology ,Hydroxylation ,01 natural sciences ,Microbiology ,Bile Acids and Salts ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Polybrominated diphenyl ethers ,RNA, Ribosomal, 16S ,medicine ,Halogenated Diphenyl Ethers ,Animals ,Homeostasis ,Metabolomics ,Intestine, Large ,Receptor ,Biotransformation ,0105 earth and related environmental sciences ,Pharmacology ,Bile acid ,Gastrointestinal Microbiome ,Articles ,biology.organism_classification ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Liver ,Dysbiosis ,Xenobiotic ,Akkermansia muciniphila ,Corn oil ,Signal Transduction - Abstract
Polybrominated diphenyl ethers (PBDEs) are persistent environmental contaminants with well characterized toxicities in host organs. Gut microbiome is increasingly recognized as an important regulator of xenobiotic biotransformation; however, little is known about its interactions with PBDEs. Primary bile acids (BAs) are metabolized by the gut microbiome into more lipophilic secondary BAs that may be absorbed and interact with certain host receptors. The goal of this study was to test our hypothesis that PBDEs cause dysbiosis and aberrant regulation of BA homeostasis. Nine-week-old male C57BL/6 conventional (CV) and germ-free (GF) mice were orally gavaged with corn oil (10 mg/kg), BDE-47 (100 μmol/kg), or BDE-99 (100 μmol/kg) once daily for 4 days (n = 3-5/group). Gut microbiome was characterized using 16S rRNA sequencing of the large intestinal content in CV mice. Both BDE-47 and BDE-99 profoundly decreased the alpha diversity of gut microbiome and differentially regulated 45 bacterial species. Both PBDE congeners increased Akkermansia muciniphila and Erysipelotrichaceae Allobaculum spp., which have been reported to have anti-inflammatory and antiobesity functions. Targeted metabolomics of 56 BAs was conducted in serum, liver, and small and large intestinal content of CV and GF mice. BDE-99 increased many unconjugated BAs in multiple biocompartments in a gut microbiota-dependent manner. This correlated with an increase in microbial 7α-dehydroxylation enzymes for secondary BA synthesis and increased expression of host intestinal transporters for BA absorption. Targeted proteomics showed that PBDEs downregulated host BA-synthesizing enzymes and transporters in livers of CV but not GF mice. In conclusion, there is a novel interaction between PBDEs and the endogenous BA-signaling through modification of the "gut-liver axis".
- Published
- 2018
12. Regulation of protein-coding gene and long noncoding RNA pairs in liver of conventional and germ-free mice following oral PBDE exposure
- Author
-
Cindy Yanfei Li and Julia Yue Cui
- Subjects
0301 basic medicine ,Male ,lcsh:Medicine ,Gene Expression ,Biochemistry ,Transcriptome ,chemistry.chemical_compound ,Mice ,Drug Metabolism ,Transcriptional regulation ,Medicine and Health Sciences ,Halogenated Diphenyl Ethers ,lcsh:Science ,Regulation of gene expression ,Multidisciplinary ,Mammalian Genomics ,Genomics ,Lipids ,Cell biology ,Nucleic acids ,Liver ,Medical Microbiology ,Enterotype ,Environmental Pollutants ,RNA, Long Noncoding ,Metabolic Pathways ,Metabolic Networks and Pathways ,Research Article ,Microbial Genomics ,Biology ,Microbiology ,03 medical and health sciences ,Genetics ,Xenobiotic Metabolism ,Animals ,Pharmacokinetics ,Gene Regulation ,Microbiome ,Non-coding RNA ,Gene ,Pharmacology ,Biology and life sciences ,lcsh:R ,Lipid metabolism ,Lipid Metabolism ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,030104 developmental biology ,Metabolism ,chemistry ,Gene Expression Regulation ,Animal Genomics ,Long non-coding RNAs ,RNA ,lcsh:Q ,Xenobiotic - Abstract
Gut microbiome communicates with the host liver to modify hepatic xenobiotic biotransformation and nutrient homeostasis. Polybrominated diphenyl ethers (PBDEs) are persistent environmental contaminants that are detected in fatty food, household dust, and human breast milk at worrisome levels. Recently, long noncoding RNAs (lncRNAs) have been recognized as novel biomarkers for toxicological responses and may regulate the transcriptional/translational output of protein-coding genes (PCGs). However, very little is known regarding to what extent the interactions between PBDEs and gut microbiome modulate hepatic lncRNAs and PCGs, and what critical signaling pathways are impacted at the transcriptomic scale. In this study, we performed RNA-Seq in livers of nine-week-old male conventional (CV) and germ-free (GF) mice orally exposed to the most prevalent PBDE congeners BDE-47 and BDE-99 (100 μmol/kg once daily for 4-days; vehicle: corn oil, 10 ml/kg), and unveiled key molecular pathways and PCG-lncRNA pairs targeted by PBDE-gut microbiome interactions. Lack of gut microbiome profoundly altered the PBDE-mediated transcriptomic response in liver, with the most prominent effect observed in BDE-99-exposed GF mice. The top pathways up-regulated by PBDEs were related to xenobiotic metabolism, whereas the top pathways down-regulated by PBDEs were in lipid metabolism and protein synthesis in both enterotypes. Genomic annotation of the differentially regulated lncRNAs revealed that majority of these lncRNAs overlapped with introns and 3'-UTRs of PCGs. Lack of gut microbiome profoundly increased the percentage of PBDE-regulated lncRNAs mapped to the 3'-UTRs of PCGs, suggesting the potential involvement of lncRNAs in increasing the translational efficiency of PCGs by preventing miRNA-3'-UTR binding, as a compensatory mechanism following toxic exposure to PBDEs. Pathway analysis of PCGs paired with lncRNAs revealed that in CV mice, BDE-47 regulated nucleic acid and retinol metabolism, as well as circadian rhythm; whereas BDE-99 regulated fatty acid metabolism. In GF mice, BDE-47 differentially regulated 19 lncRNA-PCG pairs that were associated with glutathione conjugation and transcriptional regulation. In contrast, BDE-99 up-regulated the xenobiotic-metabolizing Cyp3a genes, but down-regulated the fatty acid-metabolizing Cyp4 genes. Taken together, the present study reveals common and unique lncRNAs and PCG targets of PBDEs in mouse liver, and is among the first to show that lack of gut microbiome sensitizes the liver to toxic exposure of BDE-99 but not BDE-47. Therefore, lncRNAs may serve as specific biomarkers that differentiate various PBDE congeners as well as environmental chemical-mediated dysbiosis. Coordinate regulation of PCG-lncRNA pairs may serve as a more efficient molecular mechanism to combat against xenobiotic insult, and especially during dysbiosis-induced increase in the internal dose of toxicants.
- Published
- 2018
13. Age-Specific Regulation of Drug-Processing Genes in Mouse Liver by Ligands of Xenobiotic-Sensing Transcription Factors
- Author
-
Curtis D. Klaassen, Cindy Yanfei Li, Helen J. Renaud, and Julia Yue Cui
- Subjects
Male ,Pregnenolone Carbonitrile ,0301 basic medicine ,UGT1A6 ,Receptors, Steroid ,medicine.medical_specialty ,Polychlorinated Dibenzodioxins ,Organic Cation Transport Proteins ,Pyridines ,Receptors, Cytoplasmic and Nuclear ,Pharmaceutical Science ,Biology ,Ligands ,030226 pharmacology & pharmacy ,Gene Expression Regulation, Enzymologic ,Special Section on Pediatric Drug Disposition and Pharmacokinetics ,GSTA4 ,03 medical and health sciences ,0302 clinical medicine ,Cytochrome P-450 Enzyme System ,Downregulation and upregulation ,Internal medicine ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,RNA, Messenger ,Glucuronosyltransferase ,Receptor ,Transcription factor ,Constitutive Androstane Receptor ,Glutathione Transferase ,Pharmacology ,Regulation of gene expression ,Pregnane X receptor ,Gene Expression Profiling ,Age Factors ,Pregnane X Receptor ,CYP1A2 ,Gene Expression Regulation, Developmental ,Aldehyde Dehydrogenase ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Animals, Newborn ,Liver ,Receptors, Aryl Hydrocarbon ,Sulfotransferases - Abstract
The xenobiotic-sensing transcription factors (xeno-sensors) AhR, CAR, and PXR upregulate the expression of many drug-processing genes (DPGs) in liver. Previous studies have unveiled profound changes in the basal expression of DPGs during development; however, knowledge on the ontogeny of the inducibility of DPGs in response to pharmacological activation of xeno-sensors is still limited. The goal of this study was to investigate the age-specific regulation of DPGs by prototypical xeno-sensor ligands: 2,3,7,8-tetrachlorodibenzodioxin (TCDD) for AhR; 1,4-bis [2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP) for CAR; and pregnane-16α-carbonitrile (PCN) for PXR during mouse liver development. The basal mRNAs of most DPGs were low during neonatal age, but gradually increased to adult levels, whereas some DPGs (Cyp1a2, Cyp2b10, Cyp3a11, Gstm2, Gstm3, Papss2, and Oatp1a4) exhibited an adolescent-predominant expression pattern. The inducibility of DPGs was age-specific: 1) during neonatal age, the highest fold increase in the mRNA expression was observed for Cyp1a2, Sult5a1, and Ugt1a9 by TCDD; Cyp3a11 and Mrp2 by TCPOBOP; as well as Gstm2 and Gstm3 by PCN; 2) during adolescent age, the highest fold increase in the mRNA expression was observed for Ugt1a6 and Mrp4 by TCDD, Cyp2b10, Ugt2b34, and Ugt2b35 by TCPOBOP, as well as Gsta1, Gsta4, Sult1e1, Ugt1a1, Mrp3, and Mrp4 by PCN; 3) in adults, the highest fold increase in the mRNA expression was observed for Aldh1a1, Aldh1a7, and Ugt2b36 by TCPOBOP, as well as Papss2 and Oatp1a4 by PCN. In conclusion, the inducibility of hepatic DPGs following the pharmacological activation of xeno-sensors is age specific.
- Published
- 2015
- Full Text
- View/download PDF
14. A6 - Investigating the role of drug transporters in furosemide absorption, food-effect and elimination using a proteomics informed-mechanistic PBPK modeling approach
- Author
-
Revathi Chapa, Mayur K. Ladamor, Arzu Selen, Abdul Basit, Aarzoo Thakur, Saranjit Singh, Sheena Sharma, Cindy Yanfei Li, and Bhagwat Prasad
- Subjects
Pharmacology ,Drug ,Physiologically based pharmacokinetic modelling ,FOOD EFFECT ,Chemistry ,media_common.quotation_subject ,Pharmaceutical Science ,Furosemide ,Transporter ,Proteomics ,Biophysics ,medicine ,Pharmacology (medical) ,Pharmaceutical sciences ,Absorption (electromagnetic radiation) ,media_common ,medicine.drug - Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.