Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 223
Filter
1.
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
2.
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
3.
Front Biosci (Landmark Ed) ; 28(9): 228, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37796687

ABSTRACT

BACKGROUND: Disorders of purine metabolism are the main cause of hyperuricemia. Current drugs for the treatment of hyperuricemia usually cause a degree of cardiovascular damage. METHODS: This study aimed to investigate the therapeutic effects of Armillaria mellea fruiting body (AFB), Armillaria rhizomorph (AR) and Armillaria mellea fermentation product (after rhizomorphs removal) (AFP) on hyperuricemic mice. The hyperuricemia mouse model was established by oral administration of potassium oxonate 0.9 g⋅kg-1 and hypoxanthine 0.5 g⋅kg-1 for two weeks. Starting from the third week, the intragastric administration of the intervention drug group was as follows: Allopurinol 0.013 g⋅kg-1, AFB (3.9 and 7.8 g⋅kg-1), AR (3.9 and 7.8 g⋅kg-1), AFP (1.95 and 3.9 g⋅kg-1) once daily for 14 days. RESULTS: Results showed that AFB, AR, and AFP reduced the contents of serum uric acid, serum creatinine, and blood urea nitrogen in hyperuricemic mice and the mechanism of action might be through up-regulation of the expression levels of organic anion transporter 1/organic anion transporter 3 proteins in kidney tissue. AR and AFP both exhibited better uric acid-lowering effects than AFB, which may be due to the higher purine content of AFB. CONCLUSIONS: Armillaria mellea and its fermentation products can treat hyperuricemia by up-regulating OAT1 protein and OAT3 protein, reducing uric acid content in mice.


Subject(s)
Armillaria , Honey , Hyperuricemia , Organic Anion Transporters , Mice , Animals , Hyperuricemia/chemically induced , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Armillaria/metabolism , Kidney , Uric Acid/metabolism , Uric Acid/pharmacology , Fermentation , alpha-Fetoproteins , Organic Anion Transporters/metabolism , Organic Anion Transporters/pharmacology , Organic Anion Transporters/therapeutic use , Purines/metabolism , Purines/pharmacology , Purines/therapeutic use
4.
Drug Metab Dispos ; 51(9): 1177-1187, 2023 09.
Article in English | MEDLINE | ID: mdl-37385755

ABSTRACT

The proximal tubule plays an important role in the kidney and is a major site of drug interaction and toxicity. Analysis of kidney toxicity via in vitro assays is challenging, because only a few assays that reflect functions of drug transporters in renal proximal tubular epithelial cells (RPTECs) are available. In this study, we aimed to develop a simple and reproducible method for culturing RPTECs by monitoring organic anion transporter 1 (OAT1) as a selection marker. Culturing RPTECs in spherical cellular aggregates increased OAT1 protein expression, which was low in the conventional two-dimensional (2D) culture, to a level similar to that in human renal cortices. By proteome analysis, it was revealed that the expression of representative two proximal tubule markers was maintained and 3D spheroid culture improved the protein expression of approximately 7% of the 139 transporter proteins detected, and the expression of 2.3% of the 4,800 proteins detected increased by approximately fivefold that in human renal cortices. Furthermore, the expression levels of approximately 4,800 proteins in three-dimensional (3D) RPTEC spheroids (for 12 days) were maintained for over 20 days. Cisplatin and adefovir exhibited transporter-dependent ATP decreases in 3D RPTEC spheroids. These results indicate that the 3D RPTEC spheroids developed by monitoring OAT1 gene expression are a simple and reproducible in vitro experimental system with improved gene and protein expressions compared with 2D RPTECs and were more similar to that in human kidney cortices. Therefore, it can potentially be used for evaluating human renal proximal tubular toxicity and drug disposition. SIGNIFICANCE STATEMENT: This study developed a simple and reproducible spheroidal culture method with acceptable throughput using commercially available RPTECs by monitoring OAT1 gene expression. RPTECs cultured using this new method showed improved mRNA/protein expression profiles to those in 2D RPTECs and were more similar to those of human kidney cortices. This study provides a potential in vitro proximal tubule system for pharmacokinetic and toxicological evaluations during drug development.


Subject(s)
Kidney , Organic Anion Transport Protein 1 , Humans , Kidney/metabolism , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Kidney Tubules, Proximal/metabolism , Membrane Transport Proteins/metabolism , Gene Expression , Epithelial Cells/metabolism
5.
Biomed Pharmacother ; 160: 114342, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36739760

ABSTRACT

The Organic Anion Transporter 1 is a membrane transporter known for its central role in drug elimination by the kidney. hOAT1 is an antiporter translocating substrate in exchange for a-ketoglutarate. The understanding of hOAT1 structure and function remains limited due to the absence of resolved structure of hOAT1. Benefiting from conserved structural and functional patterns shared with other Major Facilitator Superfamily transporters, the present study intended to investigate fragments of hOAT1 transport function and modulation of its activity in order to make a step forward the understanding of its transport cycle. µs-long molecular dynamics simulation of hOAT1 were carried out suggesting two plausible binding sites for a typical substrate, adefovir, in line with experimental observations. The well-known B-like motif binding site was observed in line with previous studies. However, we here propose a new inner binding cavity which is expected to be involved in substrate translocation event. Binding modes of hOAT1 co-substrate α-ketoglutarate were also investigated suggesting that it may bind to highly conserved intracellular motifs. We here hypothesise that α-ketoglutarate may disrupt the pseudo-symmetrical intracellular charge-relay system which in turn may participate to the destabilisation of OF conformation. Investigations regarding allosteric communications along hOAT1 also suggest that substrate binding event might modulate the dynamics of intracellular charge relay system, assisted by surrounding lipids as active partners. We here proposed a structural rationalisation of transport impairments observed for two single nucleotide polymorphisms, p.Arg50His and p.Arg454Gln suggesting that the present model may be used to transport dysfunctions arising from hOAT1 mutations.


Subject(s)
Ketoglutaric Acids , Organic Anion Transport Protein 1 , Humans , Organic Anion Transport Protein 1/genetics , Membrane Transport Proteins , Lipids
6.
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
7.
Toxicol Lett ; 355: 19-30, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34748852

ABSTRACT

Organic anion transporters (OATs) belong to a subgroup of the solute carrier 22 transporter family. OATs have a central role in xenobiotic disposition affecting the toxicokinetics of its substrates and inter-individual differences in their expression, activity and function impact both toxicokinetics and toxicodynamics. Amongst OATs, OAT1 (solute carrier family 22 member 6) is involved in the urinary excretion of many xenobiotics bringing substrates into renal proximal tubular cells which can then be secreted across the apical membrane into the tubule lumen. The mycotoxin ochratoxin A has been shown to have a high affinity for OAT1, which is an important renal transporter involved in its urinary excretion. Nowadays, molecular modeling techniques are widely applied to assess protein-ligand interactions and may provide a tool to depict the mechanic of xenobiotic action be it toxicokinetics or toxicodynamics. This work provides a structured pipeline consisting of docking and molecular dynamic simulations to study OAT1-ligand interactions and the impact of OAT1 polymorphisms on such interactions. Such a computational structure-based analytical framework allowed to: i) model OAT1-substrate complex formation and depict the features correlating its sequence, structure and its capability to recruit substrates; and ii) investigate the impact of OAT1 missense mutations on substrate recruitment. Perspectives on applying such a structured pipeline to xenobiotic-metabolising enzymes are discussed.


Subject(s)
Ochratoxins/toxicity , Organic Anion Transport Protein 1/metabolism , Gene Expression Regulation/drug effects , Genetic Variation , Humans , Models, Molecular , Molecular Docking Simulation , Organic Anion Transport Protein 1/genetics , Protein Conformation , Structure-Activity Relationship
8.
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
9.
J Clin Lab Anal ; 35(8): e23859, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34251052

ABSTRACT

BACKGROUND: This study aimed to evaluate the urate-lowering effects of Yi-Suan-Cha and explore its underlying mechanisms in experimental hyperuricemia induced in rats. METHODS: Forty-eight male SD rats were randomly allocated into normal control, model, allopurinol, benzbromarone, low-dose Yi-Suan-Cha (0.2 g/ml), and high-dose Yi-Suan-Cha (0.4 g/ml) groups (n = 8 rats per group). Rat models of hyperuricemia were established through intragastric administration of adenine 25 mg/kg + potassium oxalate 300 mg/kg for 3 weeks. After the last administration, serum uric acid, creatinine, and urea nitrogen levels were measured. Renal histopathology was observed by hematoxylin-eosin staining. Xanthine oxidase level in serum and liver homogenates was measured by ELISA. The protein and mRNA expression of URAT1, ABCG2, OAT1, and GLUT9 in the kidney was detected by Western blotting and RT-PCR, respectively. RESULTS: The serum uric acid levels were significantly lowered in all medication groups than in the model group. The benzbromarone and both Yi-Suan-Cha groups showed clear kidney structures with no obvious abnormalities. Compared with the normal control group, the model group showed increased URAT1/GLUT9 protein expression and decreased ABCG2/OAT1 protein expression. Compared with the model group, both Yi-Suan-Cha groups showed decreased URAT1/GLUT9 protein expression and increased ABCG2/OAT1 protein expression. Compared with that in the normal control group, URAT1/GLUT9 mRNA expression increased in the model group. Compared with the model group, the low-dose and high-dose Yi-Suan-Cha groups showed decreased URAT1/GLUT9 mRNA expression and increased ABCG2/OAT1 mRNA expression. CONCLUSION: Yi-Suan-Cha may lower uric acid level by downregulating URAT1/GLUT9 expression and upregulating ABCG2/OAT1 expression.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Hyperuricemia/drug therapy , Kidney/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Blood Urea Nitrogen , Creatinine/blood , Disease Models, Animal , Gene Expression Regulation/drug effects , Hyperuricemia/metabolism , Hyperuricemia/pathology , Kidney/metabolism , Kidney/pathology , Male , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Rats, Sprague-Dawley , Uric Acid/blood , Xanthine Oxidase/blood , Xanthine Oxidase/metabolism
10.
Clin Genet ; 100(2): 132-143, 2021 08.
Article in English | MEDLINE | ID: mdl-33871046

ABSTRACT

Anthracyclines, chemotherapeutic agents that have contributed to significant improvements in cancer survival, also carry risk of both acute and chronic cardiotoxicity. This has led to significantly elevated risks of cardiac morbidity and mortality among cancer survivors treated with these agents. Certain treatment related, demographic, and medical factors increase an individual's risk of anthracycline induced cardiotoxicity; however, significant variability among those affected suggests that there is an underlying genetic predisposition to anthracycline induced cardiotoxicity. The current narrative review seeks to summarize the literature to date that has identified genetic variants associated with anthracycline induced cardiotoxicity. These include variants found in genes that encode proteins associated with anthracycline transportation and metabolism, those that encode proteins associated with the generation of reactive oxygen species, and those known to be associated with cardiac disease. While there is strong evidence that susceptibility to anthracycline induced cardiotoxicity has genetic underpinnings, the majority of work to date has been candidate gene analyses. Future work should focus on genome-wide analyses including genome-wide association and sequencing-based studies to confirm and expand these findings.


Subject(s)
Anthracyclines/adverse effects , Cardiomyopathies/chemically induced , Cardiomyopathies/genetics , Genetic Predisposition to Disease , ATP Binding Cassette Transporter, Subfamily B/genetics , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Antibiotics, Antineoplastic/adverse effects , Cardiomyopathies/metabolism , Cardiotoxicity/genetics , Humans , NADPH Oxidases/genetics , Nitric Oxide Synthase Type III/genetics , Organic Anion Transport Protein 1/genetics
11.
J Biol Chem ; 296: 100603, 2021.
Article in English | MEDLINE | ID: mdl-33785360

ABSTRACT

Organic anion transporter 1 (OAT1/SLC22A6) is a drug transporter with numerous xenobiotic and endogenous substrates. The Remote Sensing and Signaling Theory suggests that drug transporters with compatible ligand preferences can play a role in "organ crosstalk," mediating overall organismal communication. Other drug transporters are well known to transport lipids, but surprisingly little is known about the role of OAT1 in lipid metabolism. To explore this subject, we constructed a genome-scale metabolic model using omics data from the Oat1 knockout mouse. The model implicated OAT1 in the regulation of many classes of lipids, including fatty acids, bile acids, and prostaglandins. Accordingly, serum metabolomics of Oat1 knockout mice revealed increased polyunsaturated fatty acids, diacylglycerols, and long-chain fatty acids and decreased ceramides and bile acids when compared with wildtype controls. Some aged knockout mice also displayed increased lipid droplets in the liver when compared with wildtype mice. Chemoinformatics and machine learning analyses of these altered lipids defined molecular properties that form the structural basis for lipid-transporter interactions, including the number of rings, positive charge/volume, and complexity of the lipids. Finally, we obtained targeted serum metabolomics data after short-term treatment of rodents with the OAT-inhibiting drug probenecid to identify potential drug-metabolite interactions. The treatment resulted in alterations in eicosanoids and fatty acids, further supporting our metabolic reconstruction predictions. Consistent with the Remote Sensing and Signaling Theory, the data support a role of OAT1 in systemic lipid metabolism.


Subject(s)
Lipid Metabolism , Organic Anion Transport Protein 1/metabolism , Animals , Gene Knockout Techniques , Genomics , Machine Learning , Mice , Organic Anion Transport Protein 1/deficiency , Organic Anion Transport Protein 1/genetics
12.
Int Immunopharmacol ; 88: 106997, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33182042

ABSTRACT

Paeoniflorin-6'-O-benzene sulfonate (CP-25) is a derivative of paeoniflorin. We previously confirmed that CP-25 inhibits inflammatory responses in several arthritis animal models. The aim of the present study was to investigate the beneficial effects of CP-25 on renal damage in rats with collagen-induced arthritis (CIA). CIA was induced in rats, which were orally administered CP-25 (25, 50 and 100 mg/kg/day) for 24 days. The levels of plasma blood urea nitrogen (BUN) and urine protein in CIA rats were measured. Pathological changes in renal tissues and joints were observed, and inflammatory cell infiltration was evaluated by immunohistochemistry. Moreover, renal inflammatory mediators and transporters were measured by western blotting. We found that CP-25 not only inhibited arthritis manifestations but also improved renal pathological manifestations and kidney injury by decreasing serum BUN and urine protein levels. Further study revealed that CP-25 treatment reduced the number of renal CD68+ cells and downregulated the levels of MCP-1, TNF-α and IL-6 in CIA rats. On the other hand, we noted that CP-25 decreased the ratios of phosphorylated NF-κB p65 (p-p65) to total p65 and p-IκBα to total IκBα in CIA rats, suggesting that CP-25 blocked NF-κB activation. Finally, we observed that CP-25 restored the abnormal expression of OAT1 and OCT1 in the renal tissues of CIA rat. Our data indicate that CP-25 ameliorates kidney damage in CIA rats, and this beneficial effect is closely related to inhibiting renal inflammation and the abnormal expression of transporters.


Subject(s)
Arthritis, Experimental/complications , Glucosides/therapeutic use , Inflammation/drug therapy , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Monoterpenes/therapeutic use , Animals , Catecholamine Plasma Membrane Transport Proteins/genetics , Catecholamine Plasma Membrane Transport Proteins/metabolism , Gene Expression Regulation/drug effects , Male , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Rats , Rats, Sprague-Dawley
13.
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
14.
Molecules ; 25(13)2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32630784

ABSTRACT

Cisplatin is an anticancer drug commonly used for solid tumors. However, it causes nephrotoxicity. OAT1 and OAT3 are organic anion transporters known to contribute to the uptake of cisplatin into renal tubular cells. The present study was designed to examine the protective role of ellagic acid nanoformulation (ellagic acid nano) on cisplatin-induced nephrotoxicity in rats, and the role of OAT1/OAT3 in this effect. Four groups of male Wistar rats were used (n = 6): (1) control, (2) cisplatin (7.5 mg/kg single dose, intraperitoneal), (3) cisplatin + ellagic acid nano (1 mg/kg), and (4) cisplatin + ellagic acid nano (2 mg/kg). Nephrotoxic rats treated with ellagic acid nano exhibited a significant reduction in elevated serum creatinine, urea, and oxidative stress marker, malondialdehyde (MDA). Additionally, ellagic acid nano restored renal glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). Ellagic acid nano improved the histopathological changes induced by cisplatin, such as tubular dilatation, necrosis, and degeneration. Interestingly, OAT1 and OAT3 showed significantly lower expression at both mRNA and protein levels following ellagic acid nano treatment relative to the cisplatin-exposed group. These findings reveal a potential inhibitory role of ellagic acid antioxidant on OAT1 and OAT3 expression and thus explains its nephroprotective effect against cisplatin nephrotoxicity.


Subject(s)
Cisplatin/adverse effects , Ellagic Acid/pharmacology , Kidney/drug effects , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/administration & dosage , Cisplatin/pharmacology , Creatinine/blood , Ellagic Acid/administration & dosage , Ellagic Acid/chemistry , Female , Kidney/pathology , Male , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nanostructures/administration & dosage , Nanostructures/chemistry , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Protective Agents/pharmacology , Rats, Sprague-Dawley , Urea/blood , Xenograft Model Antitumor Assays
15.
Gene ; 752: 144790, 2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32439376

ABSTRACT

Many anatomical regions in the kidney, including proximal tubules, differ between males and females. While such differences in renal structures and functions under various physiological and pharmacological conditions have been identified, information relating to molecular mechanisms behind this gender disparity remain unknown. To understand gene expression differences in proximal tubules from human male and female kidneys, we reported on kidney cellular landscape using single-cell RNA sequencing. Differential gene expression profiles were observed in proximal tubules, between the sexes. Interestingly, the SLC22 family of anion transporters, including SLC22A6 and SLC22A8, had different expression profiles between male and female proximal tubule clusters but not sex-dependent abundance at the protein level. Moreover, in different species, we revealed a shared and species-specific differential gene expression between human and mouse kidney proximal tubules. Taken together, at single-cell resolution, this transcriptomic map represents a baseline description of gender biased genes in human kidney proximal tubules, which provide important insights for further studies of physiological differences in kidney.


Subject(s)
Kidney Tubules, Proximal/metabolism , Kidney/pathology , Sex Factors , Animals , Female , Humans , Kidney/metabolism , Kidney Tubules, Proximal/physiology , Male , Mice , Mice, Inbred C57BL , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome/genetics
16.
JCI Insight ; 5(7)2020 04 09.
Article in English | MEDLINE | ID: mdl-32271169

ABSTRACT

The role of the renal organic anion transporters OAT1 (also known as SLC22A6, originally identified as NKT) and OAT3 (also known as SLC22A8) in chronic kidney disease (CKD) remains poorly understood. This is particularly so from the viewpoint of residual proximal tubular secretion, a key adaptive mechanism to deal with protein-bound uremic toxins in CKD. Using the subtotal nephrectomy (STN) model, plasma metabolites accumulating in STN rats treated with and without the OAT inhibitor, probenecid, were identified. Comparisons with metabolomics data from Oat1-KO and Oat3-KO mice support the centrality of the OATs in residual tubular secretion of uremic solutes, such as indoxyl sulfate, kynurenate, and anthranilate. Overlapping our data with those of published metabolomics data regarding gut microbiome-derived uremic solutes - which can have dual roles in signaling and toxicity - indicates that OATs play a critical role in determining their plasma levels in CKD. Thus, the OATs, along with other SLC and ABC drug transporters, are critical to the movement of uremic solutes across tissues and into various body fluids, consistent with the remote sensing and signaling theory. The data support a role for OATs in modulating remote interorganismal and interorgan communication (gut microbiota-blood-liver-kidney-urine). The results also have implications for understanding drug-metabolite interactions involving uremic toxins.


Subject(s)
Gastrointestinal Microbiome , Kidney Tubules/metabolism , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Kidney Tubules/pathology , Mice , Mice, Knockout , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Rats , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/microbiology
17.
J Pharmacol Toxicol Methods ; 102: 106681, 2020.
Article in English | MEDLINE | ID: mdl-32087362

ABSTRACT

INTRODUCTION: This study was aimed to assess uric acid (UA)-lowering effect and its possible mechanisms of a natural complex product Yaocha in a live zebrafish model. METHODS: The zebrafish high UA model was established by feeding 5 dpf zebrafish with both an uricase inhibitor potassium oxonate at 10 mM and an UA synthesis precursor xanthine sodium at 0.5 mM for 24 h. Yaocha was administered to the high UA zebrafish through soaking at 3 various concentrations, with allopurinol as a positive control. UA level, xanthine oxidase (XOD) activity, and mRNA expression of hypoxanthine guanine-phosphoribosyltransferases transferase (HPRT1) and organic anion transporter 1 (OAT1) were measured. RESULTS: Yaocha effectively reduced UA level and inhibited xanthine oxidase (XO) activity in the high UA zebrafish. Yaocha could be a potential therapeutics for hyperuricemia through up-regulating HPRT1 and OAT1 gene expression and suppressing XO activity. DISCUSSION: These results suggested that Yaocha hold a potential for high UA prevention and therapy, possibly through inhibiting UA production and promoting urate secretion and purine conversion.


Subject(s)
Biological Products/pharmacology , Hyperuricemia/drug therapy , Uric Acid/blood , Animals , Aspalathus/chemistry , Biological Products/administration & dosage , Biological Products/chemistry , Dipeptides/administration & dosage , Dipeptides/pharmacology , Disease Models, Animal , Hypoxanthine Phosphoribosyltransferase/genetics , Organic Anion Transport Protein 1/genetics , Theaceae/chemistry , Zebrafish
18.
Clin Transl Sci ; 13(5): 848-860, 2020 09.
Article in English | MEDLINE | ID: mdl-32100958

ABSTRACT

Polymorphisms in drug transporters, like the adenosine triposphate-binding cassette (ABC) and solute carrier (SLC) superfamilies, may contribute to the observed diversity in drug response in African patients. This review aims to provide a comprehensive summary and analysis of the frequencies and distributions in African populations of ABC and SLC variants that affect drug pharmacokinetics (PK) and pharmacodynamics (PD). Of polymorphisms evaluated in African populations, SLCO1B1 rs4149056 and SLC22A6 rs1158626 were found at markedly higher frequencies than in non-African populations. SLCO1B1 rs4149056 was associated with reduction in rifampin exposure, which has implications for dosing this important anti-tuberculosis therapy. SLC22A6 rs1158626 was associated with increased affinity for antiretroviral drugs. Genetic diversity in SLC and ABC transporters in African populations has implications for conventional therapies, notably in tuberculosis and HIV. More PK and PD data in African populations are needed to assess potential for a different response to drugs compared with other global populations.


Subject(s)
Black People/genetics , Drug-Related Side Effects and Adverse Reactions/genetics , Liver-Specific Organic Anion Transporter 1/genetics , Organic Anion Transport Protein 1/genetics , Pharmacogenomic Variants , Africa/epidemiology , Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/adverse effects , Anti-Retroviral Agents/pharmacokinetics , Antitubercular Agents/administration & dosage , Antitubercular Agents/adverse effects , Antitubercular Agents/pharmacokinetics , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/prevention & control , HIV Infections/drug therapy , HIV Infections/epidemiology , Health Status Disparities , Humans , Incidence , Liver-Specific Organic Anion Transporter 1/metabolism , Organic Anion Transport Protein 1/metabolism , Pharmacogenomic Testing , Tuberculosis/drug therapy , Tuberculosis/epidemiology
19.
J Biol Chem ; 295(7): 1829-1842, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31896576

ABSTRACT

The multispecific organic anion transporters, OAT1 (SLC22A6) and OAT3 (SLC22A8), the main kidney elimination pathways for many common drugs, are often considered to have largely-redundant roles. However, whereas examination of metabolomics data from Oat-knockout mice (Oat1 and Oat3KO) revealed considerable overlap, over a hundred metabolites were increased in the plasma of one or the other of these knockout mice. Many of these relatively unique metabolites are components of distinct biochemical and signaling pathways, including those involving amino acids, lipids, bile acids, and uremic toxins. Cheminformatics, together with a "logical" statistical and machine learning-based approach, identified a number of molecular features distinguishing these unique endogenous substrates. Compared with OAT1, OAT3 tends to interact with more complex substrates possessing more rings and chiral centers. An independent "brute force" approach, analyzing all possible combinations of molecular features, supported the logical approach. Together, the results suggest the potential molecular basis by which OAT1 and OAT3 modulate distinct metabolic and signaling pathways in vivo As suggested by the Remote Sensing and Signaling Theory, the analysis provides a potential mechanism by which "multispecific" kidney proximal tubule transporters exert distinct physiological effects. Furthermore, a strong metabolite-based machine-learning classifier was able to successfully predict unique OAT1 versus OAT3 drugs; this suggests the feasibility of drug design based on knockout metabolomics of drug transporters. The approach can be applied to other SLC and ATP-binding cassette drug transporters to define their nonredundant physiological roles and for analyzing the potential impact of drug-metabolite interactions.


Subject(s)
Metabolomics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Toxins, Biological/metabolism , Adenosine Triphosphate/genetics , Animals , Bile Acids and Salts/metabolism , Biological Transport/genetics , Humans , Inactivation, Metabolic/genetics , Kidney Tubules, Proximal/metabolism , Machine Learning , Mice , Mice, Knockout , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Signal Transduction
20.
Physiol Rep ; 7(21): e14229, 2019 11.
Article in English | MEDLINE | ID: mdl-31724834

ABSTRACT

Organic anion transporter 1 (OAT1) is a polyspecific transport protein located in the basolateral membrane of renal proximal tubule cells. OAT1 plays a pivotal role in drug clearance. Adverse drug reactions (ADR) are observed more frequently in women than in men, especially ADR are higher in women for drugs which are known interactors of OAT1. Sex-dependent expression of Oat1 has been observed in rodents with a tendency to male-dominant expression. This study aims at elucidating the transcriptional regulation of human OAT1 and tests the effect of estrogen receptor α (ERα). Promoter activation of OAT1 was assessed by luciferase assays carried out by Opossum kidney (OK) cells, transiently transfected with promoter constructs of human OAT1 and expression vectors for ERα and exposed to 100 nmol/L 17ß-estradiol. Furthermore, a transcription factor array and proteomic analysis was performed to identify estrogen-induced transcription factors. Human OAT1 was significantly activated by ligand activated ERα. However, activation occurred without a direct interaction of ERα with the OAT1 promoter. Our data rather show an activation of the transcription factors CCAAT-box-binding transcription factor (CBF) and heterogeneous nuclear ribonucleoprotein K (HNRNPK) by ERα, which in turn bind and initiate OAT1 promoter activity. Herewith, we provide novel evidence of estrogen-dependent, transcriptional regulation of polyspecific drug transporters including the estrogen-induced transcription factors CBF and HNRNPK.


Subject(s)
Estrogen Receptor alpha/genetics , Gene Expression Regulation , Organic Anion Transport Protein 1/genetics , Transcription, Genetic , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cells, Cultured , Estradiol/administration & dosage , Female , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Humans , Male , Opossums , Promoter Regions, Genetic , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL
...