Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 495
Filter
1.
Pharmacol Rep ; 76(2): 400-415, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38530582

ABSTRACT

BACKGROUND: In predictions about hepatic clearance (CLH), a number of studies explored the role of albumin and transporters in drug uptake by liver cells, challenging the traditional free-drug theory. It was proposed that liver uptake can occur for transporter substrate compounds not only from the drug's unbound form but also directly from the drug-albumin complex, a phenomenon known as uptake facilitated by albumin. In contrast to albumin, dextran does not exhibit binding properties for compounds. However, as a result of its inherent capacity for stabilization, it is widely used to mimic conditions within cells. METHODS: The uptake of eight known substrates of the organic anion-transporting polypeptide 1B3 (OATP1B3) was assessed using a human embryonic kidney cell line (HEK293), which stably overexpresses this transporter. An inert polymer, dextran, was used to simulate cellular conditions, and the results were compared with experiments involving human plasma and human serum albumin (HSA). RESULTS: This study is the first to demonstrate that dextran increases compound uptake in cells with overexpression of the OATP1B3 transporter. Contrary to the common theory that highly protein-bound ligands interact with hepatocytes to increase drug uptake, the results indicate that dextran's interaction with test compounds does not significantly increase concentrations near the cell membrane surface. CONCLUSIONS: We evaluated the effect of dextran on the uptake of known substrates using OATP1B3 overexpressed in the HEK293 cell line, and we suggest that its impact on drug concentrations in liver cells may differ from the traditional role of plasma proteins and albumin.


Subject(s)
Dextrans , Organic Anion Transporters , Humans , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/pharmacology , Liver-Specific Organic Anion Transporter 1/genetics , Liver-Specific Organic Anion Transporter 1/metabolism , Liver-Specific Organic Anion Transporter 1/pharmacology , HEK293 Cells , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Hepatocytes/metabolism , Liver , Membrane Transport Proteins/metabolism , Albumins , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism
2.
Dis Markers ; 2024: 5930566, 2024.
Article in English | MEDLINE | ID: mdl-38222853

ABSTRACT

Genetic variations in urate transporters play a significant role in determining human urate levels and have been implicated in developing hyperuricemia or gout. Polymorphism in the key urate transporters, such as ABCG2, URAT1, or GLUT9 was well-documented in the literature. Therefore in this study, our objective was to determine the frequency and effect of rare nonsynonymous allelic variants of SLC22A11, SLC22A13, and SLC17A1 on urate transport. In a cohort of 150 Czech patients with primary hyperuricemia and gout, we examined all coding regions and exon-intron boundaries of SLC22A11, SLC22A13, and SLC17A1 using PCR amplification and Sanger sequencing. For comparison, we used a control group consisting of 115 normouricemic subjects. To examine the effects of the rare allelic nonsynonymous variants on the expression, intracellular processing, and urate transporter protein function, we performed a functional characterization using the HEK293A cell line, immunoblotting, fluorescent microscopy, and site directed mutagenesis for preparing variants in vitro. Variants p.V202M (rs201209258), p.R343L (rs75933978), and p.P519L (rs144573306) were identified in the SLC22A11 gene (OAT4 transporter); variants p.R16H (rs72542450), and p.R102H (rs113229654) in the SLC22A13 gene (OAT10 transporter); and the p.W75C variant in the SLC17A1 gene (NPT1 transporter). All variants minimally affected protein levels and cytoplasmic/plasma membrane localization. The functional in vitro assay revealed that contrary to the native proteins, variants p.P519L in OAT4 (p ≤ 0.05), p.R16H in OAT10 (p ≤ 0.05), and p.W75C in the NPT1 transporter (p ≤ 0.01) significantly limited urate transport activity. Our findings contribute to a better understanding of (1) the risk of urate transporter-related hyperuricemia/gout and (2) uric acid handling in the kidneys.


Subject(s)
Gout , Hyperuricemia , Organic Anion Transporters, Sodium-Independent , Organic Anion Transporters , Sodium-Phosphate Cotransporter Proteins, Type I , Humans , Gout/genetics , Hyperuricemia/genetics , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Uric Acid/metabolism , Sodium-Phosphate Cotransporter Proteins, Type I/genetics
3.
Xenobiotica ; 53(8-9): 559-571, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37885225

ABSTRACT

Cisplatin is a widely used chemotherapeutic agent to treat solid tumours in clinics. However, cisplatin-induced acute kidney injury (AKI) limits its clinical application. This study investigated the effect of hyperoside (a flavonol glycoside compound) on regulating AKI.The model of cisplatin-induced AKI was established, and hyperoside was preadministered to investigate its effect on improving kidney injury.Hyperoside ameliorated renal pathological damage, reduced the accumulation of SCr, BUN, Kim-1 and indoxyl sulphate in vivo, increased the excretion of indoxyl sulphate into the urine, and upregulated the expression of renal organic anion transporter 1 (Oat1). Moreover, evaluation of rat kidney slices demonstrated that hyperoside promoted the uptake of PAH (p-aminohippurate, the Oat1 substrate), which was confirmed by transient over-expression of OAT1 in HEK-293T cells. Additionally, hyperoside upregulated the mRNA expression of Oat1 upstream regulators hepatocyte nuclear factor-1α (HNF-1α) and pregnane X receptor (PXR).These findings indicated hyperoside could protect against cisplatin-induced AKI by promoting indoxyl sulphate excretion through regulating the expression and function of Oat1, suggesting hyperoside may offer a potential tactic for cisplatin-induced AKI treatment.


Subject(s)
Acute Kidney Injury , Cisplatin , Rats , Animals , Cisplatin/adverse effects , Cisplatin/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Indican/toxicity , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Kidney/metabolism
4.
Int J Mol Sci ; 24(20)2023 Oct 21.
Article in English | MEDLINE | ID: mdl-37895098

ABSTRACT

Organic anion transporters 1 and 3 (OAT1 and OAT3) play a crucial role in kidney function by regulating the secretion of multiple renally cleared small molecules and toxic metabolic by-products. Assessing the activity of these transporters is essential for drug development purposes as they can significantly impact drug disposition and safety. OAT1 and OAT3 are amongst the most abundant drug transporters expressed in human renal proximal tubules. However, their expression is lost when cells are isolated and cultured in vitro, which is a persistent issue across all human and animal renal proximal tubule cell models, including primary cells and cell lines. Although it is well known that the overall expression of drug transporters is affected in vitro, the underlying reasons for the loss of OAT1 and OAT3 are still not fully understood. Nonetheless, research into the regulatory mechanisms of these transporters has provided insights into the molecular pathways underlying their expression and activity. In this review, we explore the regulatory mechanisms that govern the expression and activity of OAT1 and OAT3 and investigate the physiological changes that proximal tubule cells undergo and that potentially result in the loss of these transporters. A better understanding of the regulation of these transporters could aid in the development of strategies, such as introducing microfluidic conditions or epigenetic modification inhibitors, to improve their expression and activity in vitro and to create more physiologically relevant models. Consequently, this will enable more accurate assessment for drug development and safety applications.


Subject(s)
Organic Anion Transporters, Sodium-Independent , Organic Anion Transporters , Animals , Humans , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Kidney/metabolism , Kidney Tubules, Proximal/metabolism , Epithelial Cells/metabolism , Organic Anion Transporters/metabolism
5.
J Formos Med Assoc ; 122(7): 648-652, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36964102

ABSTRACT

Rotor syndrome is a rare, benign, inherited disorder that is commonly associated with mild hyperbilirubinemia. It is caused by bi-allelic pathological variants in both SLCO1B1 and SLCO1B3 genes, causing defective OATP1B1 and OATP1B3 in the sinusoidal membrane and interrupted bilirubin uptake of the hepatocytes. We report five Taiwanese pediatric and adult patients aged 5-32 years presenting with conjugated hyperbilirubinemia, and were found to have genetic variants of SLCO1B1 and SLCO1B3. Two also had history of prolonged neonatal jaundice. Genetic analysis using panel-based next generation sequencing revealed three patients with homozygous mutations c.1738C>T (p.R580∗) in SLCO1B1 and a transposon LINE-1 insertion in SLCO1B3, one patient with homozygous mutations for another haplotype, c.757C>T (p.R253∗) in SLCO1B1 and c.1747+1G>A in SLCO1B3. Another patient had heterozygous c.1738C>T (p.R580∗) in SLCO1B1 linked with a LINE-1 insertion in SLCO1B3, and heterozygous c.757C>T (p.R253∗) in SLCO1B1 linked with c.1747+1G>A in SLCO1B3. In conclusion, we present the first time of genetic diagnosis of Rotor syndrome in Taiwan. Advanced genetic testing has enhanced the diagnosis of rare diseases with mild symptoms.


Subject(s)
Hyperbilirubinemia, Hereditary , Organic Anion Transporters , Adult , Infant, Newborn , Humans , Child , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters/genetics , Liver-Specific Organic Anion Transporter 1/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Hyperbilirubinemia, Hereditary/genetics , Hyperbilirubinemia , Mutation
6.
Toxicol Lett ; 379: 48-55, 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-36958672

ABSTRACT

Diethylene glycol (DEG) mass poisonings have resulted from ingestion of adulterated pharmaceuticals, leading to proximal tubular necrosis and acute kidney injury. Diglycolic acid (DGA), one of the primary metabolites, accumulates greatly in kidney tissue and its direct administration results in toxicity identical to that in DEG-treated rats. DGA is a dicarboxylic acid, similar in structure to Krebs cycle intermediates such as succinate. Previous studies have shown that DGA is taken into kidney cells via the succinate-related dicarboxylate transporters. These studies have assessed whether the DGA that is taken up by primary cultures of human proximal tubule (HPT) cells is effluxed. In addition, a possible mechanism for efflux, via organic anion transporters (OATs) that exchange external organic anions for dicarboxylates inside the cell, was assessed using transformed cell lines that actively express OAT activities. When HPT cells were cultured on membrane inserts, then loaded with DGA and treated with the OAT4/5 substrate estrone sulfate or the OAT1/3 substrate para-aminohippurate, no DGA efflux was seen. A repeat of this experiment utilizing RPTEC/TERT1 cells with overexpressed OAT1 and OAT3 had similar results. In these cells, but not in HPT cells, co-incubation with succinate increased the uptake of PAH, confirming the presence of OAT activity in the RPTEC/TERT1 cells. Thus, despite OATs stimulation in cells with OAT activity, there was little to no efflux of DGA from the cells. This study concluded that DGA is poorly transported out of cells and that stimulation of OAT transporters is not a viable target for reducing DGA accumulation in cells.


Subject(s)
Glycolates , Kidney Tubules, Proximal , Rats , Humans , Animals , Kidney Tubules, Proximal/metabolism , Glycolates/toxicity , Glycolates/metabolism , Succinates/metabolism , Succinic Acid/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism
7.
Cells ; 11(7)2022 03 22.
Article in English | MEDLINE | ID: mdl-35406626

ABSTRACT

The OAT1 (SLC22A6) and OAT3 (SLC22A8) urate transporters are located on the basolateral membrane of the proximal renal tubules, where they ensure the uptake of uric acid from the urine back into the body. In a cohort of 150 Czech patients with primary hyperuricemia and gout, we examined the coding regions of both genes using PCR amplification and Sanger sequencing. Variants p.P104L (rs11568627) and p.A190T (rs146282438) were identified in the gene for solute carrier family 22 member 6 (SLC22A6) and variants p.R149C (rs45566039), p.V448I (rs11568486) and p.R513Q (rs145474422) in the gene solute carrier family 22 member 8 (SLC22A8). We performed a functional study of these rare non-synonymous variants using the HEK293T cell line. We found that only p.R149C significantly reduced uric acid transport in vitro. Our results could deepen the understanding of uric acid handling in the kidneys and the molecular mechanism of uric acid transport by the OAT family of organic ion transporters.


Subject(s)
Gout , Hyperuricemia , Organic Anion Transport Protein 1 , Organic Anion Transporters, Sodium-Independent , Biological Transport , Gout/genetics , Gout/metabolism , HEK293 Cells , Humans , Hyperuricemia/genetics , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Uric Acid/metabolism
8.
Int J Mol Sci ; 23(5)2022 Feb 23.
Article in English | MEDLINE | ID: mdl-35269592

ABSTRACT

The choroid plexuses (CPs), located in the brain ventricles, form an interface between the blood and the cerebrospinal fluid named the blood-cerebrospinal barrier, which, by the presence of tight junctions, detoxification enzymes, and membrane transporters, limits the traffic of molecules into the central nervous system. It has already been shown that sex hormones regulate several CP functions, including the oscillations of its clock genes. However, it is less explored how the circadian rhythm regulates CP functions. This study aimed to evaluate the impact of sex hormones and circadian rhythms on the function of CP membrane transporters. The 24 h transcription profiles of the membrane transporters rAbca1, rAbcb1, rAbcc1, rAbcc4, rAbcg2, rAbcg4, and rOat3 were characterized in the CPs of intact male, intact female, sham-operated female, and gonadectomized rats. We found that rAbcc1 is expressed in a circadian way in the CPs of intact male rats, rAbcg2 in the CPs of intact female rats, and both rAbcc4 and rOat3 mRNA levels were expressed in a circadian way in the CPs of intact male and female rats. Next, using an in vitro model of the human blood-cerebrospinal fluid barrier, we also found that methotrexate (MTX) is transported in a circadian way across this barrier. The circadian pattern of Abcc4 found in the human CP epithelial papilloma cells might be partially responsible for MTX circadian transport across the basal membrane of CP epithelial cells.


Subject(s)
Choroid Plexus/metabolism , Methotrexate/pharmacokinetics , Multidrug Resistance-Associated Proteins/genetics , Papilloma, Choroid Plexus/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Castration , Cell Line, Tumor , Circadian Rhythm , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Methotrexate/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Papilloma, Choroid Plexus/drug therapy , Papilloma, Choroid Plexus/genetics , Rats , Sex Characteristics
9.
BMC Genom Data ; 22(1): 47, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34732138

ABSTRACT

BACKGROUND: Our preliminary work confirmed that, SLC22A7 (solute carrier family 22 member 7), NGFR (nerve growth factor receptor), ARNTL (aryl hydrocarbon receptor nuclear translocator like) and PPP2R2B (protein phosphatase 2 regulatory subunit Bß) genes were differentially expressed in dairy cows during different stages of lactation, and involved in the lipid metabolism through insulin, PI3K-Akt, MAPK, AMPK, mTOR, and PPAR signaling pathways, so we considered these four genes as the candidates affecting milk production traits. In this study, we detected polymorphisms of the four genes and verified their genetic effects on milk yield and composition traits in a Chinese Holstein cow population. RESULTS: By resequencing the whole coding region and part of the flanking region of SLC22A7, NGFR, ARNTL and PPP2R2B, we totally found 20 SNPs, of which five were located in SLC22A7, eight in NGFR, three in ARNTL, and four in PPP2R2B. Using Haploview4.2, we found three haplotype blocks including five SNPs in SLC22A7, eight in NGFR and three in ARNTL. Single-SNP association analysis showed that 19 out of 20 SNPs were significantly associated with at least one of milk yield, fat yield, fat percentage, protein yield or protein percentage in the first and second lactations (P < 0.05). Haplotype-based association analysis showed that the three haplotypes were significantly associated with at least one of milk yield, fat yield, fat percentage, protein yield or protein percentage (P < 0.05). Further, we used SOPMA software to predict a SNP, 19:g.37095131C > T in NGFR, changed the structure of NGFR protein. In addition, we used Jaspar software to found that four SNPs, 19:g.37113872C > G,19:g.37113157C > T, and 19:g.37112276C > T in NGFR and 15:g.39320936A > G in ARNTL, could change the transcription factor binding sites and might affect the expression of the corresponding genes. These five SNPs might be the potential functional mutations for milk production traits in dairy cattle. CONCLUSIONS: In summary, we proved that SLC22A7, NGFR, ARNTL and PPP2R2B have significant genetic effects on milk production traits. The valuable SNPs can be used as candidate genetic markers for genomic selection of dairy cattle, and the effects of these SNPs on other traits need to be further verified.


Subject(s)
ARNTL Transcription Factors/genetics , Cattle/genetics , Milk/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Protein Phosphatase 2/genetics , Receptors, Nerve Growth Factor/genetics , Animals , China , Female
10.
Eur J Pharmacol ; 912: 174592, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34699754

ABSTRACT

Phellodendri Chinensis Cortex (PC) is a traditional medicinal material used to treat gout and hyperuricemia (HUA) in China. Berberine (BBR), the main component of PC, possesses anti-hyperuricemic and anti-gout effects. However, BBR exhibits low bioavailability due to its extensive metabolism and limited absorption. Thus, the metabolites of BBR are believed to be the potential active forms responsible for its in vivo biological activities. Berberrubine (BRB), one of the major metabolites of BBR, exhibits appreciable biological activities even superior to BBR. In this work, the anti-hyperuricemic efficacy of BRB was investigated in HUA model mice induced by co-administration with intraperitoneal potassium oxonate (PO) and oral hypoxanthine (HX) for 7 days. Results showed that administration with BRB (6.25, 12.5, and 25.0 mg/kg) significantly decreased the serum levels of uric acid (UA) by 49.70%, 75.35%, and 75.96% respectively, when compared to the HUA group. In addition, BRB sharply decreased the levels of blood urea nitrogen (BUN) (by 19.62%, 28.98%, and 38.72%, respectively) and serum creatinine (CRE) (by 16.19%, 25.07%, and 52.08%, respectively) and reversed the PO/HX-induced renal histopathological damage dose-dependently. Additionally, BRB lowered the hepatic XOD activity, downregulated the expressions of glucose transporter 9 (GLUT9) and urate transporter 1 (URAT1), upregulated expressions of organic anion transporter 1/3 (OAT1/3) and ATP-binding cassette transporter subfamily G member 2 (ABCG2) at both protein and mRNA levels, and suppressed the activation of the JAK2/STAT3 signaling pathway. In addition, BRB significantly decreased the levels of inflammatory mediators (IL-1ß, IL-6, and TNF-α). In conclusion, our study indicated that BRB exerted anti-hyperuricemic effect, at least in part, via regulating the urate transporter expressions and suppressing the JAK2/STAT3 signaling pathway. BRB was believed to be promising for further development into a potential therapeutic agent for HUA treatment.


Subject(s)
Berberine/analogs & derivatives , Hyperuricemia/drug therapy , Janus Kinase 2/metabolism , Organic Anion Transporters/metabolism , Protective Agents/pharmacology , STAT3 Transcription Factor/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Berberine/pharmacology , Berberine/therapeutic use , Blood Urea Nitrogen , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Creatinine/blood , Cytokines/metabolism , Disease Models, Animal , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Hyperuricemia/chemically induced , Hypoxanthine/toxicity , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Male , Mice , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Oxonic Acid/toxicity , Protective Agents/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Uric Acid/blood , Xanthine Oxidase/metabolism
11.
PLoS One ; 16(5): e0250408, 2021.
Article in English | MEDLINE | ID: mdl-33945567

ABSTRACT

Gyps species have been previously shown to be highly sensitive to the toxic effects of diclofenac, when present in their food sources as drug residues following use as a veterinary medicine. Vultures exposed to diclofenac soon become depressed and die with signs of severe visceral gout and renal damage on necropsy. The molecular mechanism behind toxicity and renal excretion of uric acid is still poorly understood. With the clinical pictures suggesting renal uric acid excretion as the target site for toxicity, as a first step the following study was undertaken to determine the uric acid excretory pathways present in the African white-backed vulture (Gyps africanus) (AWB), one of the species susceptible to toxicity. Using transcriptome analysis, immunohistochemistry and functional predictions, we demonstrated that AWB makes use of the organic anion transporter 2 (OAT2) for their uric acid excretion. RT-qPCR analysis subsequently demonstrated relatively similar expression of the OAT2 transporter in the vulture and chicken. Lastly docking analysis, predicted that the non-steroidal drugs induce their toxicity through an allosteric binding.


Subject(s)
Avian Proteins/metabolism , Kidney/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , Birds , Organic Anion Transporters, Sodium-Independent/chemistry , Organic Anion Transporters, Sodium-Independent/genetics , Transcriptome , Uric Acid/metabolism
12.
Biochem Pharmacol ; 188: 114546, 2021 06.
Article in English | MEDLINE | ID: mdl-33838133

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world. The failure of chemotherapy in HCC patients is partly due to inadequate intracellular drug accumulation caused by abnormally expressed drug transporters. Human organic anion transporter 2 (hOAT2), a transporter mainly expressed in liver and kidney, is responsible for uptake of various antineoplastic drugs such as 5-fluorouracil (5-FU). Among 32 pairs of human HCC samples, we preliminarily found that OAT2 was suppressed in HCC tissues compared with matched tumor-adjacent tissues at both mRNA and protein levels, which resulted in 5-FU resistance in HCC. However, the epigenetic regulatory mechanisms of OAT2 downregulation have not been investigated. In this study, we first proved it was histone hypoacetylation rather than DNA hypermethylation that participated in transcriptional repression of OAT2 in two HCC cell lines (BEL-7402 and SMMC-7721). In general, there were two pathways confirmed using tissues and cells: 1) Increased histone deacetylase sirtuin 7 (SIRT7) mediated loss of histone 3 lysine 18 acetylation (H3K18ac) at the promoter of OAT2 and inhibited its transcription. 2) More histone deacetylase 7 (HDAC7) instead of lysine acetyltransferase 8 (KAT8) enrichment at the promoter of OAT2 led to low levels of histone 4 lysine 16 acetylation (H4K16ac). Further, we found that histone deacetylases inhibitor vorinostat (SAHA) could reverse histone hypoacetylation state to activate OAT2 transcription and enhance uptake of classic OAT2 substrate zidovudine. Therefore, we evaluated the effect of combining SAHA and 5-FU and the results demonstrated that SAHA could sensitize HCC cells to 5-FU. Collectively, we proposed such a combination treatment to overcome 5-FU resistance in HCC from the perspective of epigenetically restoring OAT2.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Fluorouracil/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/biosynthesis , Liver Neoplasms/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Acetylation/drug effects , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Dose-Response Relationship, Drug , Fluorouracil/therapeutic use , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Histone Deacetylases/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/genetics , Up-Regulation/drug effects , Up-Regulation/physiology , Vorinostat/pharmacology
13.
J Pharmacol Sci ; 146(1): 21-28, 2021 May.
Article in English | MEDLINE | ID: mdl-33858651

ABSTRACT

Paeoniflorin-6'-O-benzene sulfonate (CP-25) is a derivative of Paeoniflorin. We investigate beneficial effect of CP-25 on methotrexate (MTX) induced nephrotoxicity in rats. Plasma blood urea nitrogen (Bun), plasma creatinine (CREA), urine CREA and protein in the rats were quantitatively measured. Renal tissues were pathologically observed, and apoptosis was detected. Apoptosis related proteins and organic anion transporter-3 (OAT3) expression were determined by western blotting analysis. MTX induced nephrotoxicity and hematotoxicity in rats with abnormal levels of serum Bun, serum CERA, 24 h urine protein excretion, white blood cells, platelets, plateletcrit and abnormal renal pathological appearance. Either pre-treatment or treatment of CP-25 restored normal levels of hematological and renal function parameters, and improved histopathology in rats treated with MTX. CP-25 prevented MTX induced apoptosis of renal tubular cells, and the effect was further confirmed by its regulatory effects on abnormal expression of Bax, cleaved-caspase-3, cleaved-caspase-8, Cyt-c, Bcl-2. The other important finding is co-administration of CP-25 with MTX significantly increased MTX renal excretion in the damaged rats, and the effect is supposed to be linked with its regulation on abnormal renal OAT3 expression. Taken together, CP-25 shows well protective activity against MTX induced nephrotoxicity, and this effect is via its anti-apoptosis and detoxification properties.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Apoptosis/drug effects , Apoptosis/genetics , Glucosides/pharmacology , Glucosides/therapeutic use , Kidney/drug effects , Kidney/metabolism , Methotrexate/adverse effects , Methotrexate/metabolism , Monoterpenes/pharmacology , Monoterpenes/therapeutic use , Acute Kidney Injury/metabolism , Animals , Disease Models, Animal , Gene Expression/drug effects , Gene Expression/genetics , Kidney/pathology , Male , Molecular Targeted Therapy , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Rats, Sprague-Dawley
14.
Ann Rheum Dis ; 80(9): 1220-1226, 2021 09.
Article in English | MEDLINE | ID: mdl-33832965

ABSTRACT

OBJECTIVES: To perform a genome-wide association study (GWAS) of gout cases versus asymptomatic hyperuricaemia (AH) controls, and gout cases versus normouricaemia controls, and to generate a polygenic risk score (PRS) to determine gout-case versus AH-control status. METHODS: Gout cases and AH controls (serum urate (SU) ≥6.0 mg/dL) from the UK Biobank were divided into discovery (4934 cases, 56 948 controls) and replication (2115 cases, 24 406 controls) cohorts. GWAS was conducted and PRS generated using summary statistics in discovery cohort as the base dataset and the replication cohort as the target dataset. The predictive ability of the model was evaluated. GWAS were performed to identify variants associated with gout compared with normouricaemic controls using SU <6.0 mg/dL and <7.0 mg/dL thresholds, respectively. RESULTS: Thirteen independent single nucleotide polymorphisms (SNPs) in ABCG2, SLC2A9, SLC22A11, GCKR, MEPE, PPM1K-DT, LOC105377323 and ADH1B reached genome-wide significance and replicated as predictors of AH to gout transition. Twelve of 13 associations were novel for this transition, and rs1229984 (ADH1B) was identified as GWAS locus for gout for the first time. The best PRS model was generated from association data of 17 SNPs; and had predictive ability of 58.5% that increased to 69.2% on including demographic factors. Two novel SNPs rs760077(MTX1) and rs3800307(PRSS16) achieved GWAS significance for association with gout compared with normouricaemic controls using both SU thresholds. CONCLUSION: The association of urate transporters with gout supports the central role of hyperuricaemia in its pathogenesis. Larger GWAS are required to identify if variants in inflammatory pathways contribute to progression from AH to gout.


Subject(s)
Asymptomatic Diseases , Gout/genetics , Hyperuricemia/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Adaptor Proteins, Signal Transducing/genetics , Aged , Alcohol Dehydrogenase/genetics , Disease Progression , Extracellular Matrix Proteins/genetics , Female , Genome-Wide Association Study , Glucose Transport Proteins, Facilitative/genetics , Glycoproteins/genetics , Humans , Logistic Models , Male , Middle Aged , Mitochondrial Membrane Transport Proteins/genetics , Multifactorial Inheritance , Neoplasm Proteins/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Phosphoproteins/genetics , Polymorphism, Single Nucleotide , Risk Factors , Serine Endopeptidases/genetics , White People
15.
Ecotoxicol Environ Saf ; 211: 111910, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33444879

ABSTRACT

Cadmium (Cd) is a typical pollutant and carcinogen in environment. Exposure assessment of contaminants is an important component of occupational and environmental epidemiological studies. Early studies of Cd have focused on aquatic animals, chickens and rats. However, toxicological evaluation of Cd in pigs has not been reported. Therefore, twelve pigs were randomly divided into two groups (n = 6): the control group and the Cd group (Cd content: 15 ± 0.242 mg/kg feed) in this study, the experimental period was 30 d, and the toxic effects of Cd on the liver of weanling piglets were examined by antioxidant function, liver function, Cd content, histological examination and transcriptomics. The results showed that the changes of antioxidant function, liver function and Cd content were significant in the liver. Transcriptional profiling results showed that 399 differentially expressed genes (DEGs) were significantly up-regulated while 369 DEGs were remarkably down-regulated in Cd group, and which were concentrated in three ontologies: molecular function, cellular component and biological processes. Interestingly, significant changes in some genes of the cytochrome P450 enzyme (CYP450) and solute carrier (SLC) families have been observed and were consistent with qRT-PCR results. In conclusion, Cd could cause liver injury in weanling piglets and change the transcriptomic characteristics of liver. CYP450 and SLC families play an indispensable role in Cd-mediated hepatotoxicity. Importantly, changes in mRNA levels of CYP2B22, CYP7A1, CYP8B1, SLC26A8, SLC11A1, SLC27A2 and SLC22A7 induced by Cd have been reported for the first time. Our findings will provide a new insight for better assessing the mechanism of Cd toxicity to the liver.


Subject(s)
Cadmium/toxicity , Chemical and Drug Induced Liver Injury, Chronic/enzymology , Cytochrome P-450 Enzyme System/metabolism , Environmental Pollutants/toxicity , Animals , Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury, Chronic/pathology , Liver/drug effects , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/pharmacology , RNA, Messenger/genetics , Swine , Transcriptome/drug effects
16.
Poult Sci ; 99(12): 6371-6377, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33248552

ABSTRACT

We previously reported that blue eggshell color in chickens is associated with a partial endogenous retroviral (EAV-HP) insertion in the promoter region of the solute carrier organic anion transporter family member 1B3 (SLCO1B3) gene. The EAV-HP sequence includes numerous regulatory elements, which may modulate the expression of adjacent genes. To determine whether this insertion influences the expression of neighboring genes, we screened the expression of solute carrier organic anion transporter family members 1C1, 1B1 (SLCO1C1, SLCO1B1), and SLCO1B3 in 13 and 10 tissues from female and male Yimeng chickens, respectively. We observed that the insertion only significantly modulated the expression of SLCO1B3 and did not majorly affect that of SLCO1C1 and SLCO1B1. High expression of SLCO1B3 was detected in the shell gland, magnum, isthmus, and vagina of the oviduct in female blue-eggshell chickens. We also observed ectopic expression of SLCO1B3 in the testes of male chickens. SLCO1B3 is typically highly expressed in the liver; however, the EAV-HP insertion significantly reduces SLCO1B3 expression. As a liver-specific transporter, a reduction in the expression of SLCO1B3 may affect liver metabolism, particularly that of bile acids. We also detected higher ectopic expression of SLCO1B3 in the lungs of birds heterozygous for the EAV-HP insertion than in homozygous genotypes. In conclusion, we confirmed that the EAV-HP insertion modifies SLCO1B3 expression, and showed, for the first time, similar expression profile of this gene in all parts of the oviduct in females and testis in males. We also observed different levels of SLCO1B3 expression in the liver, which were associated with the EAV-HP insertion, and significantly higher expression in the lungs of birds with heterozygous genotype. The effects of these changes in the SLCO1B3 expression pattern on the function of the tissues warrant further investigation.


Subject(s)
5' Flanking Region , Chickens , Egg Shell , Endogenous Retroviruses , Gene Expression , Organic Anion Transporters, Sodium-Independent , 5' Flanking Region/genetics , Animals , Chickens/genetics , Chickens/metabolism , Egg Shell/metabolism , Endogenous Retroviruses/genetics , Female , Male , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Ovum/metabolism , Pigmentation/genetics
17.
Article in English | MEDLINE | ID: mdl-33013684

ABSTRACT

Bone production, maintenance, and modeling are a well-balanced process involving mineralization by osteoblasts and resorption by osteoclasts. Sex steroid hormones, including their conjugated forms, contribute majorly to maintaining this balance. Recently, variants in the SLC22A9 gene have been associated with osteoporosis in Korean females. We had recently shown that SLC22A9, encoding organic anion transporter 7 (OAT7), is an uptake transporter of estrone sulfate and identified several genetic variants in Europeans leading to functional consequences in vitro. We therefore hypothesized that SLC22A9 genetic variants may contribute to the pathophysiology of osteoporosis in Europeans. To test this hypothesis, we examined the associations of SLC22A9 variants with bone quality, fractures, and bone turnover markers. We genotyped SLC22A9 variants in 5,701 (2,930 female) subjects (age range, 20-93 years) extracted from the population-based Study of Health in Pomerania (SHIP and SHIP-TREND) covered by the Illumina Infinium HumanExome BeadChip version v1.0 (Exome Chip). Descriptive data (e.g., history of fractures), ultrasonography of the calcaneus, as well as serum concentrations of carboxy-terminal telopeptide of type I collagen, amino-terminal propeptide of type I procollagen, and vitamin D were determined. Comprehensive statistical analyses revealed no association between low-frequency and rare SLC22A9 variants and bone quality, fractures, and bone turnover markers. Our results indicate that single genetic SLC22A9 variants do not have a major impact on osteoporosis risk prediction in Europeans, yet findings need to be replicated in larger-scale studies.


Subject(s)
Biomarkers/blood , Fractures, Bone/genetics , Genetic Predisposition to Disease , Organic Anion Transporters, Sodium-Independent/genetics , Osteoporosis/epidemiology , Polymorphism, Single Nucleotide , White People/genetics , Adult , Aged , Aged, 80 and over , Bone Density , Bone Remodeling , Case-Control Studies , Cross-Sectional Studies , Europe/epidemiology , Female , Fractures, Bone/blood , Gene Expression Profiling , Humans , Male , Middle Aged , Organic Anion Transporters, Sodium-Independent/blood , Osteoporosis/blood , Osteoporosis/genetics , Osteoporosis/pathology , Prognosis , Young Adult
18.
BMC Genomics ; 21(1): 680, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32998709

ABSTRACT

BACKGROUND: Ergot alkaloids (E+) are mycotoxins produced by the endophytic fungus, Epichloë coenophiala, in tall fescue that are associated with ergotism in animals. Exposure to ergot alkaloids during gestation reduces fetal weight and placental mass in sheep. These reductions are related to vasoconstrictive effects of ergot alkaloids and potential alterations in nutrient transport to the fetus. Cotyledon samples were obtained from eight ewes that were fed E+ (n = 4; E+/E+) or E- (endophyte-free without ergot alkaloids; n = 4; E-/E-) seed during both mid (d 35 to 85) and late (d 85-133) gestation to assess differentially expressed genes associated with ergot alkaloid induced reductions in placental mass and fetal weight, and discover potential adaptive mechanisms to alter nutrient supply to fetus. RESULTS: Ewes fed E+/E+ fescue seed during both mid and late gestation had 20% reduction in fetal body weight and 33% reduction in cotyledon mass compared to controls (E-/E-). Over 13,000 genes were identified with 110 upregulated and 33 downregulated. Four genes had a |log2FC| > 5 for ewes consuming E+/E+ treatment compared to controls: LECT2, SLC22A9, APOC3, and MBL2. REViGO revealed clusters of upregulated genes associated glucose, carbohydrates, lipid, protein, macromolecular and cellular metabolism, regulation of wound healing and response to starvation. For downregulated genes, no clusters were present, but all enriched GO terms were associated with anion and monocarboxylic acid transport. The complement and coagulation cascade and the peroxisome proliferator-activated receptor signaling pathway were found to be enriched for ewes consuming E+/E+ treatment. CONCLUSIONS: Consumption of ergot alkaloids during gestation altered the cotyledonary transcriptome specifically related to macronutrient metabolism, wound healing and starvation. These results show that ergot alkaloid exposure upregulates genes involved in nutrient metabolism to supply the fetus with additional substrates in attempts to rescue fetal growth.


Subject(s)
Ergot Alkaloids/toxicity , Mycotoxins/toxicity , Placenta/metabolism , Sheep/microbiology , Transcriptome , Animal Feed/microbiology , Animals , Apolipoprotein C-III/genetics , Apolipoprotein C-III/metabolism , Epichloe/metabolism , Epichloe/pathogenicity , Female , Festuca/microbiology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Placenta/drug effects , Pregnancy , Sheep/genetics , Sheep/metabolism
19.
Toxicol Sci ; 178(2): 338-346, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32946573

ABSTRACT

In artisanal and small-scale gold mining, occupational exposure to mercury (Hg) vapor is related to harmful effects on several organs, including the kidneys. We previously reported significantly increased levels of Hg in blood and urine despite normal kidney function in individuals from Colombia occupationally exposed to Hg compared with those nonexposed. We evaluated the contribution of 4 genetic variants in key genes encoding the transporters solute carrier (SLC; rs4149170 and rs4149182) and ATP-binding cassette(ABC; rs1202169 and rs1885301) in the pathogenesis of nephrotoxicity due to Hg exposure in these groups. Regression analysis was performed to determine the association between the blood- and urine-Hg concentration with SLC and ABC polymorphisms in 281 Colombian individuals (160 exposed and 121 nonexposed to Hg). We found an enrichment of ABCB1 rs1202169-T allele in the exposed group (p = .011; OR= 2.05; 95% CI = 1.18-3.58) compared with the nonexposure group. We also found that carriers of SLC22A8 rs4149182-G and ABCB1 rs1202169-T alleles had a higher urinary clearance rate of Hg than noncarriers (ß = 0.13, p = .04), whereas carriers of SLC22A6 rs4149170-A and ABCB1 rs1202169-C alleles showed abnormal levels of estimated glomerular filtration rate (ß = -84.96, p = .040) and beta-2-microglobulin (ß = 743.38, p < .001). Our results suggest that ABCB1 rs1202169 and its interaction with SLC22A8 rs4149182 and SLC22A6 rs4149170 could mitigate Hg nephrotoxicity by controlling the renal proximal tubule cell accumulation of inorganic Hg. This will be useful to estimate the risk of kidney toxicity associated to Hg and the genetic selection to aid adaptation to Hg-rich environments.


Subject(s)
Mercury , Mining , Organic Anion Transporters, Sodium-Independent/genetics , Polymorphism, Genetic , ATP Binding Cassette Transporter, Subfamily B/genetics , Adolescent , Adult , Colombia , Environmental Monitoring , Female , Gold , Humans , Male , Mercury/toxicity , Middle Aged , Organic Anion Transport Protein 1/genetics , Young Adult
20.
Pharm Res ; 37(8): 158, 2020 Aug 02.
Article in English | MEDLINE | ID: mdl-32743772

ABSTRACT

PURPOSE: Drug elimination alteration has been well reported in acute lymphoblastic leukemia (ALL). Considering that transporters and glomerular filtration influence, to different extents, the drug disposition, and possible side effects, we evaluated the effects of ALL on major renal transporters and glomerular filtration mediated pharmacokinetic changes, as well as expression of renal drug transporters. METHODS: ALL xenograft models were established and intravenously injected with substrates of renal transporters and glomerular filtration separately in NOD/SCID mice. The plasma concentrations of substrates, after single doses, were determined using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). RESULTS: With the development of ALL, protein expression of MDR1, OAT3 and OCT2 were increased by 2.62-fold, 1.70-fold, and 1.45-fold, respectively, whereas expression of MRP2 and MRP4 were significantly decreased by 30.98% and 45.28% in the kidney of ALL groups compared with control groups. Clearance of MDR1-mediated digoxin, OAT3-mediated furosemide, and OCT2-mediated metformin increased by 3.04-fold, 1.47-fold, and 1.26-fold, respectively. However, clearance of MRPs-mediated methotrexate was reduced by 39.5%. These results are consistent with mRNA expression. Clearance of vancomycin and amikacin, as markers of glomerular filtration rate, had a 2.14 and 1.64-fold increase in ALL mice, respectively. CONCLUSIONS: The specific alteration of renal transporters and glomerular filtration in kidneys provide a rational explanation for changes in pharmacokinetics for ALL.


Subject(s)
Glomerular Filtration Rate/physiology , Kidney/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Renal Elimination/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Chromatography, High Pressure Liquid , Digoxin/administration & dosage , Digoxin/pharmacokinetics , Furosemide/administration & dosage , Furosemide/pharmacokinetics , Gene Expression Regulation , Humans , Male , Metformin/administration & dosage , Metformin/pharmacokinetics , Mice, Inbred NOD , Mice, SCID , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...