Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.215
Filter
1.
Biol Pharm Bull ; 47(9): 1484-1486, 2024.
Article in English | MEDLINE | ID: mdl-39231688

ABSTRACT

Membrane transporters expressed in the choroid plexus (CP) are involved in the transport of substances between the blood and cerebrospinal fluid (CSF). Carnitine/organic cation transporter 1 (OCTN1, also known as SLC22A4) is expressed in rodent CP; however, its specific roles in blood-CSF transport remain unclear. Therefore, in this study, we aimed to evaluate the potential role of OCTN1 in the elimination of substances from CSF. Tritium-labeled ergothioneine ([3H]ERGO), a typical in vivo substrate of OCTN1, was injected into the lateral ventricles of wild-type and octn1 gene knockout (octn1-/-) mice. Clearance of [3H]ERGO from CSF was higher than that of the bulk flow marker, [14C]mannitol, in wild-type mice. However, [3H]ERGO clearance was significantly lower in octn1-/- mice than in wild-type mice. Furthermore, OCTN1 expression in CP was determined via immunohistochemical analysis. CP/CSF ratio of [3H]ERGO was significantly lower in octn1-/- mice than in wild-type mice. These results suggest that OCTN1 is functionally expressed in CP and involved in the elimination of ERGO from CSF in mice.


Subject(s)
Choroid Plexus , Ergothioneine , Mice, Knockout , Organic Cation Transport Proteins , Animals , Choroid Plexus/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Ergothioneine/metabolism , Ergothioneine/cerebrospinal fluid , Mice , Male , Mice, Inbred C57BL , Symporters
2.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126116

ABSTRACT

Plantaginis semen is the dried ripe seed of Plantago asiatica L. or Plantago depressa Willd., which has a long history in alleviating hyperuricemia (HUA) and chronic kidney diseases. While the major chemical ingredients and mechanism remained to be illustrated. Therefore, this work aimed to elucidate the chemicals and working mechanisms of PS for HUA. UPLC-QE-Orbitrap-MS was applied to identify the main components of PS in vitro and in vivo. RNA sequencing (RNA-seq) was conducted to explore the gene expression profile, and the genes involved were further confirmed by real-time quantitative PCR (RT-qPCR). A total of 39 components were identified from PS, and 13 of them were detected in the rat serum after treating the rat with PS. The kidney tissue injury and serum uric acid (UA), xanthine oxidase (XOD), and cytokine levels were reversed by PS. Meanwhile, renal urate anion transporter 1 (Urat1) and glucose transporter 9 (Glut9) levels were reversed with PS treatment. RNA-seq analysis showed that the PPAR signaling pathway; glycine, serine, and threonine metabolism signaling pathway; and fatty acid metabolism signaling pathway were significantly modified by PS treatment. Further, the gene expression of Slc7a8, Pck1, Mgll, and Bhmt were significantly elevated, and Fkbp5 was downregulated, consistent with RNA-seq results. The PPAR signaling pathway involved Pparα, Pparγ, Lpl, Plin5, Atgl, and Hsl were elevated by PS treatment. URAT1 and PPARα proteins levels were confirmed by Western blotting. In conclusion, this study elucidates the chemical profile and working mechanisms of PS for prevention and therapy of HUA and provides a promising traditional Chinese medicine agency for HUA prophylaxis.


Subject(s)
Hyperuricemia , Oxonic Acid , Plantago , Hyperuricemia/drug therapy , Hyperuricemia/metabolism , Animals , Rats , Oxonic Acid/adverse effects , Male , Plantago/chemistry , Uric Acid/blood , Plant Extracts/pharmacology , Kidney/metabolism , Kidney/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Organic Anion Transporters/metabolism , Organic Anion Transporters/genetics , Xanthine Oxidase/metabolism
3.
Int J Mol Sci ; 25(16)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39201429

ABSTRACT

OCTN1 and OCTN2 are membrane transport proteins encoded by the SLC22A4 and SLC22A5 genes, respectively. Even though several transcripts have been predicted by bioinformatics for both genes, only one functional protein isoform has been described for each of them. Both proteins are ubiquitous, and depending on the physiopathological state of the cell, their expression is regulated by well-known transcription factors, although some aspects have been neglected. A plethora of missense variants with uncertain clinical significance are reported both in the dbSNP and the Catalogue of Somatic Mutations in Cancer (COSMIC) databases for both genes. Due to their involvement in human pathologies, such as inflammatory-based diseases (OCTN1/2), systemic primary carnitine deficiency (OCTN2), and drug disposition, it would be interesting to predict the impact of variants on human health from the perspective of precision medicine. Although the lack of a 3D structure for these two transport proteins hampers any speculation on the consequences of the polymorphisms, the already available 3D structures for other members of the SLC22 family may provide powerful tools to perform structure/function studies on WT and mutant proteins.


Subject(s)
Gene Expression Regulation , Solute Carrier Family 22 Member 5 , Humans , Solute Carrier Family 22 Member 5/genetics , Solute Carrier Family 22 Member 5/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/chemistry , Protein Conformation , Symporters/genetics , Symporters/metabolism , Symporters/chemistry
4.
Food Chem Toxicol ; 192: 114949, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39182635

ABSTRACT

Acute kidney injury (AKI) is a worldwide public health problem with high morbidity and mortality. Cisplatin is a widely used chemotherapeutic agent for treating solid tumors, but the induction of AKI restricts its clinical application. In this study, the effect of cisplatin on the expression of organic ion transporters was investigated through in vivo and in vitro experiments. Targeted metabolomics techniques were used to measure the levels of selected endogenous substances in serum. Transmission electron microscopy was used to observe the microstructure of renal tubular epithelial cells. Our results show that the toxicity of cisplatin on HK-2 cells or HEK-293 cells was time- and dose-dependent. Administration of cisplatin decreased the expression of OAT1/3 and OCT2 and increased the expression of MRP2/4. Mitochondrial damage induced by cisplatin lead to renal tubular epithelial cell injury. In addition, administration of cisplatin resulted in significant changes in endogenous substance levels in serum, including amino acids, carnitine, and fatty acids. These serum amino acids and metabolites (α-aminobutyric acid, proline, and alanine), carnitines (tradecanoylcarnitine, hexanylcarnitine, octanoylcarnitine, 2-methylbutyroylcarnitine, palmitoylcarnitine, and linoleylcarnitine) and fatty acids (9E-tetradecenoic acid) represent endogenous substances with diagnostic potential for cisplatin-induced AKI.


Subject(s)
Acute Kidney Injury , Cisplatin , Cisplatin/toxicity , Humans , Animals , HEK293 Cells , Male , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Kidney/drug effects , Kidney/metabolism , Antineoplastic Agents/toxicity , Organic Anion Transporters/metabolism , Organic Anion Transporters/genetics , Organic Cation Transporter 2/metabolism , Organic Cation Transporter 2/genetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Carnitine/analogs & derivatives , Carnitine/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism
5.
Endocrinol Diabetes Metab ; 7(5): e486, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39086121

ABSTRACT

BACKGROUND: The response of patients with Type 2 diabetes mellitus (T2DM) to metformin may be a variation because of genetic differences in solute carrier (SLC) transporter proteins and other effect factors, which have an important effect on how metformin is processed in the body and its efficiency for glycaemic control. AIM: This study was conducted to investigate the impact of certain genetic variants of the organic cation transporter genes OCT3 (SLC22A3 rs12194182 and rs8187722) and MATE2 (SLC47A2 rs12943590) and their association with glycaemic parameters in patients with T2DM who respond poorly to metformin. PATIENTS AND METHODS: This cross-sectional study involved 150 Iraqi cases with T2DM who were prescribed a daily dose of (1000 mg/day) metformin for a minimum of 3 months. Various parameters included are as follows: demographic data, glycaemic parameters and three SNPs: rs12943590 variant of SLC47A2, rs12194182 and rs8187722 variant of SLC22A3 using the standard PCR-sequencing technique. RESULTS: Thirty-nine patients (26.17%) were responders, whereas 111 patients (73.82%) could not respond to metformin treatment. Upon analysing the genotypes of the rs12943590 variants of SLC47A2, rs12194182 and rs8187722 SNPs of SLC22A3, the present findings revealed a nonsignificant association of genetic variations in all SNPs with metformin response. SLC47A2 (rs12943590) showed nonsignificant associations of the GG, AA and AG genotyping; SLC22A3 (rs12194182) showed nonsignificant associations of the TT, TC and CC genotyping; and SLC22A3 (rs8187722) showed nonsignificant associations of the AA, CC and AC genotyping between two groups. CONCLUSION: Variations in genes SLC22A3 and SLC47A2 did not have a significant role in the response of patients with T2DM to metformin (1000 mg/day).


Subject(s)
Diabetes Mellitus, Type 2 , Hypoglycemic Agents , Metformin , Organic Cation Transport Proteins , Polymorphism, Single Nucleotide , Humans , Metformin/administration & dosage , Metformin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Organic Cation Transport Proteins/genetics , Male , Female , Middle Aged , Cross-Sectional Studies , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Adult , Aged , Genotype , Blood Glucose
6.
Cells ; 13(12)2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38920639

ABSTRACT

The polarised expression of specific transporters in proximal tubular epithelial cells is important for the renal clearance of many endogenous and exogenous compounds. Thus, ideally, the in vitro tools utilised for predictions would have a similar expression of apical and basolateral xenobiotic transporters as in vivo. Here, we assessed the functionality of organic cation and anion transporters in proximal tubular-like cells (PTL) differentiated from human induced pluripotent stem cells (iPSC), primary human proximal tubular epithelial cells (PTEC), and telomerase-immortalised human renal proximal tubular epithelial cells (RPTEC/TERT1). Organic cation and anion transport were studied using the fluorescent substrates 4-(4-(dimethylamino)styryl)-N-methylpyridinium iodide (ASP) and 6-carboxyfluorescein (6-CF), respectively. The level and rate of intracellular ASP accumulation in PTL following basolateral application were slightly lower but within a 3-fold range compared to primary PTEC and RPTEC/TERT1 cells. The basolateral uptake of ASP and its subsequent apical efflux could be inhibited by basolateral exposure to quinidine in all models. Of the three models, only PTL showed a modest preferential basolateral-to-apical 6-CF transfer. These results show that organic cation transport could be demonstrated in all three models, but more research is needed to improve and optimise organic anion transporter expression and functionality.


Subject(s)
Epithelial Cells , Kidney Tubules, Proximal , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/cytology , Epithelial Cells/metabolism , Models, Biological , Pyridinium Compounds/metabolism , Anions/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Biological Transport , Organic Anion Transporters/metabolism , Organic Anion Transporters/genetics , Cell Line , Cations/metabolism , Fluoresceins/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics
7.
PLoS One ; 19(6): e0304512, 2024.
Article in English | MEDLINE | ID: mdl-38829838

ABSTRACT

The Organic Cation Transporter Novel 1 (OCTN1), also known as SLC22A4, is widely expressed in various human tissues, and involved in numerous physiological and pathological processes remains. It facilitates the transport of organic cations, zwitterions, with selectivity for positively charged solutes. Ergothioneine, an antioxidant compound, and acetylcholine (Ach) are among its substrates. Given the lack of experimentally solved structures of this protein, this study aimed at generating a reliable 3D model of OCTN1 to shed light on its substrate-binding preferences and the role of sodium in substrate recognition and transport. A chimeric model was built by grafting the large extracellular loop 1 (EL1) from an AlphaFold-generated model onto a homology model. Molecular dynamics simulations revealed domain-specific mobility, with EL1 exhibiting the highest impact on overall stability. Molecular docking simulations identified cytarabine and verapamil as highest affinity ligands, consistent with their known inhibitory effects on OCTN1. Furthermore, MM/GBSA analysis allowed the categorization of substrates into weak, good, and strong binders, with molecular weight strongly correlating with binding affinity to the recognition site. Key recognition residues, including Tyr211, Glu381, and Arg469, were identified through interaction analysis. Ach demonstrated a low interaction energy, supporting the hypothesis of its one-directional transport towards to outside of the membrane. Regarding the role of sodium, our model suggested the involvement of Glu381 in sodium binding. Molecular dynamics simulations of systems at increasing levels of Na+ concentrations revealed increased sodium occupancy around Glu381, supporting experimental data associating Na+ concentration to molecule transport. In conclusion, this study provides valuable insights into the 3D structure of OCTN1, its substrate-binding preferences, and the role of sodium in the recognition. These findings contribute to the understanding of OCTN1 involvement in various physiological and pathological processes and may have implications for drug development and disease management.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Organic Cation Transport Proteins , Humans , Organic Cation Transport Proteins/chemistry , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Symporters/chemistry , Symporters/metabolism , Binding Sites , Protein Binding , Ergothioneine/chemistry , Ergothioneine/metabolism , Sodium/metabolism , Sodium/chemistry , Computer Simulation , Acetylcholine/metabolism , Acetylcholine/chemistry , Ligands
8.
Biochem Biophys Res Commun ; 726: 150269, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-38909533

ABSTRACT

Mitochondrial dysfunction is implicated in a wide range of human disorders including many neurodegenerative and cardiovascular diseases, metabolic diseases, cancers, and respiratory disorders. Studies have suggested the potential of l-ergothioneine (ET), a unique dietary thione, to prevent mitochondrial damage and improve disease outcome. Despite this, no studies have definitively demonstrated uptake of ET into mitochondria. Moreover, the expression of the known ET transporter, OCTN1, on the mitochondria remains controversial. In this study, we utilise mass spectrometry to demonstrate direct ET uptake in isolated mitochondria as well as its presence in mitochondria isolated from ET-treated cells and animals. Mitochondria isolated from OCTN1 knockout mice tissues, have impaired but still detectable ET uptake, raising the possibility of alternative transporter(s) which may facilitate ET uptake into the mitochondria. Our data confirm that ET can enter mitochondria, providing a basis for further work on ET in the prevention of mitochondrial dysfunction in human disease.


Subject(s)
Ergothioneine , Mice, Knockout , Mitochondria , Ergothioneine/metabolism , Ergothioneine/pharmacology , Animals , Mitochondria/metabolism , Humans , Mice , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Symporters/metabolism , Symporters/genetics
9.
Biosci Rep ; 44(7)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38896441

ABSTRACT

Lipoprotein(a) (Lp(a)) is a low-density lipoprotein (LDL)-like particle in which the apolipoprotein B component is covalently linked to apolipoprotein(a) (apo(a)). Lp(a) is a well-established independent risk factor for cardiovascular diseases. Plasma Lp(a) concentrations vary enormously between individuals and ethnic groups. Several nucleotide polymorphisms in the SLC22A3 gene associate with Lp(a) concentration in people of different ethnicities. We investigated the association of a Polynesian-specific (Maori and Pacific peoples) SLC22A3 gene coding variant p.Thr44Met) with the plasma concentration of Lp(a) in a cohort of 302 healthy Polynesian males. An apo(a)-size independent assay assessed plasma Lp(a) concentrations; all other lipid and apolipoprotein concentrations were measured using standard laboratory techniques. Quantitative real-time polymerase chain reaction was used to determine apo(a) isoforms. The range of metabolic (HbA1c, blood pressure, and blood lipids) and blood lipid variables were similar between the non-carriers and carriers in age, ethnicity and BMI adjusted models. However, rs8187715 SLC22A3 variant was significantly associated with lower Lp(a) concentrations. Median Lp(a) concentration was 10.60 nmol/L (IQR: 5.40-41.00) in non-carrier group, and was 7.60 nmol/L (IQR: 5.50-12.10) in variant carrier group (P<0.05). Lp(a) concentration inversely correlated with apo(a) isoform size. After correction for apo(a) isoform size, metabolic parameters and ethnicity, the association between the SLC22A3 variant and plasma Lp(a) concentration remained. The present study is the first to identify the association of this gene variant and low plasma Lp(a) concentrations. This provides evidence for better guidance on ethnic specific cut-offs when defining 'elevated' and 'normal' plasma Lp(a) concentrations in clinical applications.


Subject(s)
Apoprotein(a) , Lipoprotein(a) , Organic Cation Transport Proteins , Polymorphism, Single Nucleotide , Adult , Aged , Humans , Male , Middle Aged , Apoprotein(a)/genetics , Apoprotein(a)/blood , Lipoprotein(a)/blood , Lipoprotein(a)/genetics , Organic Cation Transport Proteins/genetics , Pacific Island People , Protein Isoforms/genetics , Protein Isoforms/blood
10.
Yakugaku Zasshi ; 144(6): 659-674, 2024.
Article in Japanese | MEDLINE | ID: mdl-38825475

ABSTRACT

Serum urate levels are determined by the balance between uric acid production and uric acid excretion capacity from the kidneys and intestinal tract. Dysuricemia, including hyperuricemia and hypouricemia, develops when the balance shifts towards an increase or a decrease in the uric acid pool. Hyperuricemia is mostly a multifactorial genetic disorder involving several disease susceptibility genes and environmental factors. Hypouricemia, on the other hand, is caused by genetic abnormalities. The main genes involved in dysuricemia are xanthine oxidoreductase, an enzyme that produces uric acid, and the urate transporters urate transporter 1/solute carrier family 22 member 12 (URAT1/SLC22A12), glucose transporter 9/solute carrier family 2 member 9 (GLUT9/SLC2A9) and ATP binding cassette subfamily G member 2 (ABCG2). Deficiency of xanthine oxidoreductase results in xanthinuria, a rare disease with marked hypouricemia. Xanthinuria can be due to a single deficiency of xanthine oxidoreductase or in combination with aldehyde oxidase deficiency as well. The latter is caused by a deficiency in molybdenum cofactor sulfurase, which is responsible for adding sulphur atoms to the molybdenum cofactor required for xanthine oxidoreductase and aldehyde oxidase to exert their action. URAT1/SLC22A12 and GLUT9/SLC2A9 are involved in urate reabsorption and their deficiency leads to renal hypouricemia, a condition that is common in Japanese due to URAT1/SLC22A12 deficiency. On the other hand, ABCG2 is involved in the secretion of urate, and many Japanese have single nucleotide polymorphisms that result in its reduced function, leading to hyperuricemia. In particular, severe dysfunction of ABCG2 leads to hyperuricemia with reduced extrarenal excretion.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2 , Glucose Transport Proteins, Facilitative , Hyperuricemia , Neoplasm Proteins , Organic Anion Transporters , Uric Acid , Xanthine Dehydrogenase , Humans , Hyperuricemia/etiology , Hyperuricemia/metabolism , Hyperuricemia/genetics , Uric Acid/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Organic Anion Transporters/metabolism , Organic Anion Transporters/genetics , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transport Proteins, Facilitative/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Xanthine Dehydrogenase/metabolism , Xanthine Dehydrogenase/genetics , Xanthine Dehydrogenase/deficiency , Animals , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/etiology , Renal Tubular Transport, Inborn Errors/metabolism , Urinary Calculi/etiology , Urinary Calculi/metabolism , Urinary Calculi/genetics , Metabolism, Inborn Errors
11.
Stroke ; 55(6): 1650-1659, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38738428

ABSTRACT

BACKGROUND: Beyond neuronal injury, cell death pathways may also contribute to vascular injury after stroke. We examined protein networks linked to major cell death pathways and identified SLC22A17 (solute carrier family 22 member 17) as a novel mediator that regulates endothelial tight junctions after ischemia and inflammatory stress. METHODS: Protein-protein interactions and brain enrichment analyses were performed using STRING, Cytoscape, and a human tissue-specific expression RNA-seq database. In vivo experiments were performed using mouse models of transient focal cerebral ischemia. Human stroke brain tissues were used to detect SLC22A17 by immunostaining. In vitro experiments were performed using human brain endothelial cultures subjected to inflammatory stress. Immunostaining and Western blot were used to assess responses in SLC22A17 and endothelial tight junctional proteins. Water content, dextran permeability, and electrical resistance assays were used to assess edema and blood-brain barrier (BBB) integrity. Gain and loss-of-function studies were performed using lentiviral overexpression of SLC22A17 or short interfering RNA against SLC22A17, respectively. RESULTS: Protein-protein interaction analysis showed that core proteins from apoptosis, necroptosis, ferroptosis, and autophagy cell death pathways were closely linked. Among the 20 proteins identified in the network, the iron-handling solute carrier SLC22A17 emerged as the mediator enriched in the brain. After cerebral ischemia in vivo, endothelial expression of SLC22A17 increases in both human and mouse brains along with BBB leakage. In human brain endothelial cultures, short interfering RNA against SLC22A17 prevents TNF-α (tumor necrosis factor alpha)-induced ferroptosis and downregulation in tight junction proteins and disruption in transcellular permeability. Notably, SLC22A17 could repress the transcription of tight junctional genes. Finally, short interfering RNA against SLC22A17 ameliorates BBB leakage in a mouse model of focal cerebral ischemia. CONCLUSIONS: Using a combination of cell culture, human stroke samples, and mouse models, our data suggest that SLC22A17 may play a role in the control of BBB function after cerebral ischemia. These findings may offer a novel mechanism and target for ameliorating BBB injury and edema after stroke.


Subject(s)
Blood-Brain Barrier , Brain Ischemia , Tight Junctions , Aged , Animals , Female , Humans , Male , Mice , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/genetics , Cell Death , Endothelial Cells/metabolism , Mice, Inbred C57BL , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Tight Junctions/metabolism
12.
Nat Commun ; 15(1): 4380, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782905

ABSTRACT

SLC22A10 is an orphan transporter with unknown substrates and function. The goal of this study is to elucidate its substrate specificity and functional characteristics. In contrast to orthologs from great apes, human SLC22A10, tagged with green fluorescent protein, is not expressed on the plasma membrane. Cells expressing great ape SLC22A10 orthologs exhibit significant accumulation of estradiol-17ß-glucuronide, unlike those expressing human SLC22A10. Sequence alignments reveal a proline at position 220 in humans, which is a leucine in great apes. Replacing proline with leucine in SLC22A10-P220L restores plasma membrane localization and uptake function. Neanderthal and Denisovan genomes show proline at position 220, akin to modern humans, indicating functional loss during hominin evolution. Human SLC22A10 is a unitary pseudogene due to a fixed missense mutation, P220, while in great apes, its orthologs transport sex steroid conjugates. Characterizing SLC22A10 across species sheds light on its biological role, influencing organism development and steroid homeostasis.


Subject(s)
Primates , Animals , Humans , Amino Acid Sequence , Estradiol/metabolism , HEK293 Cells , Hominidae/genetics , Hominidae/metabolism , Mutation, Missense , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Primates/genetics , Pseudogenes , Substrate Specificity
13.
Structure ; 32(8): 1150-1164.e3, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38815577

ABSTRACT

Multidrug and toxin extrusion (MATE) family transporters excrete toxic compounds coupled to Na+/H+ influx. Although structures of MATE transporters are available, the mechanism by which substrate export is coupled to ion influx remains unknown. To address this issue, we conducted a structural analysis of Pyrococcus furiosus MATE (PfMATE) using solution nuclear magnetic resonance (NMR). The NMR analysis, along with thorough substitutions of all non-exposed acidic residues, confirmed that PfMATE is under an equilibrium between inward-facing (IF) and outward-facing (OF) conformations, dictated by the Glu163 protonation. Importantly, we found that only the IF conformation exhibits a mid-µM affinity for substrate recognition. In contrast, the OF conformation exhibited only weak mM substrate affinity, suitable for releasing substrate to the extracellular side. These results indicate that PfMATE is an affinity-directed H+ antiporter where substrates selectively bind to the protonated IF conformation in the equilibrium, and subsequent proton release mechanistically ensures H+-coupled substrate excretion by the transporter.


Subject(s)
Archaeal Proteins , Pyrococcus furiosus , Pyrococcus furiosus/metabolism , Archaeal Proteins/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Protein Binding , Substrate Specificity , Binding Sites , Models, Molecular , Protons , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/chemistry , Organic Cation Transport Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation
14.
J Pharmacol Exp Ther ; 390(1): 99-107, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38670801

ABSTRACT

Dotinurad was developed as a uricosuric agent, inhibiting urate (UA) reabsorption through the UA transporter URAT1 in the kidneys. Due to its high selectivity for URAT1 among renal UA transporters, we investigated the mechanism underlying this selectivity by identifying dotinurad binding sites specific to URAT1. Dotinurad was docked to URAT1 using AutoDock4, utilizing the AlphaFold2-predicted structure. The inhibitory effects of dotinurad on wild-type and mutated URAT1 at the predicted binding sites were assessed through URAT1-mediated [14C]UA uptake in Xenopus oocytes. Nine amino acid residues in URAT1 were identified as dotinurad-binding sites. Sequence alignment with UA-transporting organic anion transporters (OATs) revealed that H142 and R487 were unique to URAT1 among renal UA-transporting OATs. For H142, IC50 values of dotinurad increased to 62, 55, and 76 nM for mutated URAT1 (H142A, H142E, and H142R, respectively) compared with 19 nM for the wild type, indicating that H142 contributes to URAT1-selective interaction with dotinurad. H142 was predicted to interact with the phenyl-hydroxyl group of dotinurad. The IC50 of the hydroxyl group methylated dotinurad (F13141) was 165 µM, 8420-fold higher than dotinurad, suggesting the interaction of H142 and the phenyl-hydroxyl group by forming a hydrogen bond. Regarding R487, URAT1-R487A exhibited a loss of activity. Interestingly, the URAT1-H142A/R487A double mutant restored UA transport activity, with the IC50 value of dotinurad for the mutant (388 nM) significantly higher than that for H142A (73.5 nM). These results demonstrate that H142 and R487 of URAT1 determine its selectivity for dotinurad, a uniqueness observed only in URAT1 among UA-transporting OATs. SIGNIFICANCE STATEMENT: Dotinurad selectively inhibits the urate reabsorption transporter URAT1 in renal urate-transporting organic ion transporters (OATs). This study demonstrates that dotinurad interacts with H142 and R487 of URAT1, located in the extracellular domain and unique among OATs when aligning amino acid sequences. Mutations in these residues reduce affinity of dotinurad for URAT1, confirming their role in conferring selective inhibition. Additionally, the interaction between dotinurad and URAT1 involving H142 is found to mediate hydrogen bonding.


Subject(s)
Organic Anion Transporters , Uric Acid , Uricosuric Agents , Animals , Organic Anion Transporters/metabolism , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/genetics , Uric Acid/metabolism , Uric Acid/pharmacology , Binding Sites , Humans , Uricosuric Agents/pharmacology , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/antagonists & inhibitors , Xenopus laevis , Kidney/metabolism , Kidney/drug effects , Oocytes/metabolism , Oocytes/drug effects , Benzothiazoles/pharmacology , Molecular Docking Simulation
15.
Biomolecules ; 14(4)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38672410

ABSTRACT

Inflammation is a physiological condition characterized by a complex interplay between different cells handled by metabolites and specific inflammatory-related molecules. In some pathological situations, inflammation persists underlying and worsening the pathological state. Over the years, two membrane transporters namely OCTN1 (SLC22A4) and OCTN2 (SLC22A5) have been shown to play specific roles in inflammation. These transporters form the OCTN subfamily within the larger SLC22 family. The link between these proteins and inflammation has been proposed based on their link to some chronic inflammatory diseases such as asthma, Crohn's disease (CD), and rheumatoid arthritis (RA). Moreover, the two transporters show the ability to mediate the transport of several compounds including carnitine, carnitine derivatives, acetylcholine, ergothioneine, and gut microbiota by-products, which have been specifically associated with inflammation for their anti- or proinflammatory action. Therefore, the absorption and distribution of these molecules rely on the presence of OCTN1 and OCTN2, whose expression is modulated by inflammatory cytokines and transcription factors typically activated by inflammation. In the present review, we wish to provide a state of the art on OCTN1 and OCTN2 transport function and regulation in relationships with inflammation and inflammatory diseases focusing on the metabolic signature collected in different body districts and gene polymorphisms related to inflammatory diseases.


Subject(s)
Inflammation , Organic Cation Transport Proteins , Solute Carrier Family 22 Member 5 , Symporters , Humans , Inflammation/metabolism , Solute Carrier Family 22 Member 5/metabolism , Solute Carrier Family 22 Member 5/genetics , Animals , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Ergothioneine/metabolism , Crohn Disease/metabolism , Crohn Disease/genetics , Crohn Disease/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/genetics , Gastrointestinal Microbiome , Carnitine/metabolism , Asthma/metabolism , Asthma/genetics , Acetylcholine/metabolism
16.
Biochem Pharmacol ; 223: 116188, 2024 May.
Article in English | MEDLINE | ID: mdl-38580166

ABSTRACT

Recently published cryo-EM structures of human organic cation transporters of the SLC22 family revealed seven, sequentially arranged glutamic and aspartic acid residues, which may be relevant for interactions with positively charged substrates. We analyzed the functional consequences of removing those negative charges by creating D155N, E232Q, D382N, E390Q, E451Q, E459Q, and D478N mutants of OCT3. E232Q, E459Q, and D478N resulted in a lack of localization in the outer cell membrane and no relevant uptake activity. However, D155N and E451Q showed a substrate-specific loss of transport activity, whereas E390Q had no remaining activity despite correct membrane localization. In contrast, D382N showed almost wild-type-like uptake. D155 is located at the entrance to the substrate binding pocket and could, therefore be involved in guiding cationic substrates towards the inside of the binding pocket. For E390, we confirm its critical function for transporter function as it was recently shown for the corresponding position in OCT1. Interestingly, E451 seems to be located at the bottom of the binding pocket in the outward-open confirmation of the transporter. Substrate-specific loss of transport activity of the E451Q variant suggests an essential role in the transport cycle of specific substances as part of an opportunistic binding site. In general, our study highlights the impact of the cryo-EM structures in guiding mutagenesis studies to understand the molecular level of transporter-ligand interactions, and it also confirms the importance of testing multiple substrates in mutagenesis studies of polyspecific OCTs.


Subject(s)
Amino Acids , Organic Cation Transport Proteins , Humans , Cations/metabolism , Mutagenesis , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/metabolism , Organic Cation Transporter 2
17.
Biochem Biophys Res Commun ; 712-713: 149922, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38626531

ABSTRACT

We previously reported that solute carrier family 22 member 18 (Slc22a18) regulates lipid accumulation in 3T3-L1 adipocytes. Here, we provide additional evidence derived from experiments with adenoviral vector expression and genetic manipulation of mice. In primary cultured rat hepatocytes, adenoviral overexpression of mouse Slc22a18 increased triglyceride accumulation and triglyceride synthetic activity, which was decreased in an adenoviral knockdown experiment. Adenoviral overexpression of mouse Slc22a18 in vivo caused massive fatty liver in mice, even under normal dietary conditions. Conversely, adenoviral knockdown of mouse Slc22a18 reduced hepatic lipid accumulation induced by a high-glucose and high-sucrose diet. We created Slc22a18 knockout mice, which grew normally and showed no obvious spontaneous phenotypes. However, compared with control littermates, the knockout mice exhibited decreased hepatic triglyceride content under refeeding conditions, significantly reduced epididymal fat mass, and tended to have lower liver weight in conjunction with leptin deficiency. Finally, we created transgenic mice overexpressing rat Slc22a18 in an adipose-specific manner, which had increased body weight and epididymal fat mass primarily because of increased adipocyte cell volume. In these transgenic mice, a positive correlation was observed between adiposity and the expression levels of the rat Slc22a18 transgene. Taken together, these results indicate that Slc22a18 has positive effects on lipid accumulation in vivo.


Subject(s)
Organic Cation Transport Proteins , Animals , Mice , Rats , Male , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Mice, Knockout , Hepatocytes/metabolism , Triglycerides/metabolism , Mice, Transgenic , Lipid Metabolism/genetics , Fatty Liver/metabolism , Fatty Liver/genetics , Fatty Liver/pathology , Mice, Inbred C57BL , Liver/metabolism , Adiposity/genetics , Adipocytes/metabolism , Adipose Tissue/metabolism , Cells, Cultured , Rats, Sprague-Dawley
18.
Sci Rep ; 14(1): 3925, 2024 02 16.
Article in English | MEDLINE | ID: mdl-38366023

ABSTRACT

Solute carrier family (SLC) transporters are expressed in the digestive system and play important roles in maintaining physiological functions in the body. In addition, SLC transporters act as oncoproteins or tumor-suppressor proteins during the development, progression, and metastasis of various digestive system cancers. SLC22A18, a member of the SLC22 gene family, is an orphan transporter with an unknown endogenous substrate. Previous study revealed that SLC22A18 is downregulated in colorectal cancer tissues and that it acts as a suppressor in colorectal cancer, although the effects of SLC22A18 variants on colon cancer cell proliferation, migration, and invasion are unknown. Therefore, in this study, we identified SLC22A18 variants found in multiple populations by searching public databases and determined the in vitro effects of these missense variations on transporter expression and cancer progression. Our results indicated that three missense SLC22A18 variants-p.Ala6Thr, p.Arg12Gln, and p.Arg86His-had significantly lower cell expression than the wild type, possibly owing to intracellular degradation. Furthermore, these three variants caused significantly higher proliferation, migration, and invasion of colon cancer cells than the wild type. Our findings suggest that missense variants of SLC22A18 can potentially serve as biomarkers or prognostic tools that enable clinicians to predict colorectal cancer progression.


Subject(s)
Colonic Neoplasms , Organic Cation Transport Proteins , Humans , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Prognosis , Tumor Suppressor Proteins/genetics , Organic Cation Transport Proteins/genetics
19.
Drug Metab Dispos ; 52(4): 296-304, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38326034

ABSTRACT

Accurate predictions of renal drug-drug interactions (DDIs) mediated by the human organic cation transporter 2 (hOCT2) and multidrug and toxin extrusion proteins (hMATEs) remain challenging. Current DDI evaluation using plasma maximal unbound inhibitor concentrations (Imax,u) and IC50 values determined in single transporter-transfected cells frequently leads to false or overprediction especially for hMATE1. Emerging evidence suggests intracellular unbound inhibitor concentration may be more relevant for hMATE1 inhibition in vivo. However, determination of intrarenal inhibitor concentrations is impractical. Here, we explored the use of hOCT2/hMATE1 double-transfected Madin-Darby canine kidney (MDCK) cells as a new in vitro tool for DDI risk assessment. Our results showed that potent in vitro hMATE1 inhibitors (hydroxychloroquine, brigatinib, and famotidine) failed to inhibit metformin B-to-A flux in the double-transfected system. On the other side, the classic hOCT2/hMATE1 inhibitors, pyrimethamine and cimetidine, dose-dependently inhibited metformin apparent B-to-A permeability (Papp). The different behaviors of these hMATE1 inhibitors in the double-transfected system can be explained by their different ability to gain intracellular access either via passive diffusion or transporter-mediated uptake. A new parameter (IC50,flux) was proposed reflecting the inhibitor's potency on overall hOCT2/hMATE1-mediated tubular secretion. The IC50,flux values significantly differ from the IC50 values determined in single transporter-transfected cells. Importantly, the IC50,flux accurately predicted in vivo DDIs (within 2-fold) when used in a static model. Our data demonstrated that the IC50,flux approach circumvents the need to measure intracellular inhibitor concentrations and more accurately predicted hOCT2/hMATE1-mediated renal DDIs. This system represents a new approach that could be used for improved DDI assessment during drug development. SIGNIFICANCE STATEMENT: This study demonstrated that flux studies in double-transfected MDCK cells and the IC50,flux represents a better approach to assess in vivo DDI potential for the renal organic cation secretion system. This study highlights the importance of inhibitor intracellular accessibility for accurate prediction of hMATE1-mediated renal DDIs. This approach has the potential to identify in vitro hMATE1 inhibitors that are unlikely to result in in vivo DDIs, thus reducing the burden of unnecessary and costly clinical DDI investigations.


Subject(s)
Metformin , Organic Cation Transport Proteins , Animals , Dogs , Humans , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Metformin/metabolism , Drug Interactions , Kidney/metabolism
20.
Toxicology ; 503: 153757, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38364893

ABSTRACT

Doxorubicin (DOX) is a widely used antitumor agent; however, its clinical application is limited by dose-related organ damage. Because organic cation/carnitine transporters (OCTN1 and OCTN2), which are critical for DOX uptake, are highly expressed in hepatocytes, we aimed to elucidate the role of these transporters in hepatic DOX uptake. The results indicated that inhibitors and RNA interference both significantly reduced DOX accumulation in HepG2 and HepaRG cells, suggesting that OCTN1/2 contribute substantially to DOX uptake by hepatocytes. To determine whether metformin (MET, an inhibitor of OCTN1 and OCTN2) ameliorates DOX-induced hepatotoxicity, we conducted in vitro and in vivo studies. MET (1-100 µM) inhibited DOX (500 nM) accumulation and cytotoxicity in vitro in a concentration-dependent manner. Furthermore, intravenous MET administration at 250 or 500 mg/kg or by gavage at 50, 100, or 200 mg/kg reduced DOX (8 mg/kg) accumulation in a dose-dependent manner in the mouse liver and attenuated the release of alanine aminotransferase, aspartate aminotransferase, and carboxylesterase 1. Additionally, MET reduced the distribution of DOX in the heart, liver, and kidney and enhanced the urinary elimination of DOX; however, it did not increase the nephric toxicity of DOX. In conclusion, our study demonstrated that MET alleviates DOX hepatotoxicity by inhibiting OCTN1- and OCTN2-mediated DOX uptake in vitro (mouse hepatocytes and HepaRG or HepG2 cells) and in mice.


Subject(s)
Chemical and Drug Induced Liver Injury , Metformin , Symporters , Mice , Animals , Organic Cation Transport Proteins/genetics , Solute Carrier Family 22 Member 5 , Metformin/pharmacology , Doxorubicin/toxicity , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL