1
|
Kerhoas M, Le Vée M, Carteret J, Jouan E, Tastet V, Bruyère A, Huc L, Fardel O. Inhibition of human drug transporter activities by succinate dehydrogenase inhibitors. CHEMOSPHERE 2024; 358:142122. [PMID: 38663675 DOI: 10.1016/j.chemosphere.2024.142122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 04/20/2024] [Accepted: 04/21/2024] [Indexed: 05/02/2024]
Abstract
Succinate dehydrogenase inhibitors (SDHIs) are widely-used fungicides, to which humans are exposed and for which putative health risks are of concern. In order to identify human molecular targets for these environmental chemicals, the interactions of 15 SDHIs with activities of main human drug transporters implicated in pharmacokinetics were investigated in vitro. 5/15 SDHIs, i.e., benzovindiflupyr, bixafen, fluxapyroxad, pydiflumetofen and sedaxane, were found to strongly reduce activity of the renal organic anion transporter (OAT) 3, in a concentration-dependent manner (with IC50 values in the 1.0-3.9 μM range), without however being substrates for OAT3. Moreover, these 5/15 SDHIs decreased the membrane transport of estrone-3 sulfate, an endogenous substrate for OAT3, and sedaxane was predicted to inhibit in vivo OAT3 activity in response to exposure to the acceptable daily intake (ADI) dose. In addition, pydiflumetofen strongly inhibited the renal organic cation transporter (OCT) 2 (IC50 = 2.0 μM) and benzovindiflupyr the efflux pump breast cancer resistance protein (BCRP) (IC50 = 3.9 μM). Other human transporters, including organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 as well as multidrug and toxin extrusion protein (MATE) 1 and MATE2-K were moderately or weakly inhibited by SDHIs, whereas P-glycoprotein, multidrug resistance-associated protein (MRP), OCT1 and OAT1 activities were not or only marginally impacted. Then, some human drug transporters, especially OAT3, constitute molecular targets for SDHIs. This could have toxic consequences, notably with respect to levels of endogenous compounds and metabolites substrates for the considered transporters or to potential SDHI-drug interactions. This could therefore contribute to putative health risk of these fungicides.
Collapse
Affiliation(s)
- Marie Kerhoas
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France
| | - Marc Le Vée
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France
| | - Jennifer Carteret
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France
| | - Elodie Jouan
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France
| | - Valentin Tastet
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France
| | - Laurence Huc
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France; Laboratoire Interdisciplinaire Sciences Innovations Sociétés (LISIS), INRAE/CNRS/Université Gustave Eiffel, F-Marne-La-Vallée, France
| | - Olivier Fardel
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
2
|
Dou T, Lian T, Shu S, He Y, Jiang J. The substrate and inhibitor binding mechanism of polyspecific transporter OAT1 revealed by high-resolution cryo-EM. Nat Struct Mol Biol 2023; 30:1794-1805. [PMID: 37845412 PMCID: PMC11406556 DOI: 10.1038/s41594-023-01123-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023]
Abstract
Organic anion transporters (OATs) of the SLC22 family have crucial roles in the transport of organic anions, including metabolites and therapeutic drugs, and in transporter-mediated drug-drug interactions. In the kidneys, OATs facilitate the elimination of metabolic waste products and xenobiotics. However, their transport activities can lead to the accumulation of certain toxic compounds within cells, causing kidney damage. Moreover, OATs are important drug targets, because their inhibition modulates the elimination or retention of substrates linked to diseases. Despite extensive research on OATs, the molecular basis of their substrate and inhibitor binding remains poorly understood. Here we report the cryo-EM structures of rat OAT1 (also known as SLC22A6) and its complexes with para-aminohippuric acid and probenecid at 2.1, 2.8 and 2.9 Å resolution, respectively. Our findings reveal a highly conserved substrate binding mechanism for SLC22 transporters, wherein four aromatic residues form a cage to accommodate the polyspecific binding of diverse compounds.
Collapse
Affiliation(s)
- Tongyi Dou
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Tengfei Lian
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Shi Shu
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Yi He
- Fermentation Facility, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Jiansen Jiang
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
3
|
Hussain SI, Muhammad N, Shah SUD, Fardous F, Khan SA, Khan N, Rehman AU, Siddique M, Wasan SA, Niaz R, Ullah H, Khan N, Muhammad N, Mirza MU, Wasif N, Khan S. Structural and functional implications of SLC13A3 and SLC9A6 mutations: an in silico approach to understanding intellectual disability. BMC Neurol 2023; 23:353. [PMID: 37794328 PMCID: PMC10548666 DOI: 10.1186/s12883-023-03397-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/20/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Intellectual disability (ID) is a condition that varies widely in both its clinical presentation and its genetic underpinnings. It significantly impacts patients' learning capacities and lowers their IQ below 70. The solute carrier (SLC) family is the most abundant class of transmembrane transporters and is responsible for the translocation of various substances across cell membranes, including nutrients, ions, metabolites, and medicines. The SLC13A3 gene encodes a plasma membrane-localized Na+/dicarboxylate cotransporter 3 (NaDC3) primarily expressed in the kidney, astrocytes, and the choroid plexus. In addition to three Na + ions, it brings four to six carbon dicarboxylates into the cytosol. Recently, it was discovered that patients with acute reversible leukoencephalopathy and a-ketoglutarate accumulation (ARLIAK) carry pathogenic mutations in the SLC13A3 gene, and the X-linked neurodevelopmental condition Christianson Syndrome is caused by mutations in the SLC9A6 gene, which encodes the recycling endosomal alkali cation/proton exchanger NHE6, also called sodium-hydrogen exchanger-6. As a result, there are severe impairments in the patient's mental capacity, physical skills, and adaptive behavior. METHODS AND RESULTS Two Pakistani families (A and B) with autosomal recessive and X-linked intellectual disorders were clinically evaluated, and two novel disease-causing variants in the SLC13A3 gene (NM 022829.5) and the SLC9A6 gene (NM 001042537.2) were identified using whole exome sequencing. Family-A segregated a novel homozygous missense variant (c.1478 C > T; p. Pro493Leu) in the exon-11 of the SLC13A3 gene. At the same time, family-B segregated a novel missense variant (c.1342G > A; p.Gly448Arg) in the exon-10 of the SLC9A6 gene. By integrating computational approaches, our findings provided insights into the molecular mechanisms underlying the development of ID in individuals with SLC13A3 and SLC9A6 mutations. CONCLUSION We have utilized in-silico tools in the current study to examine the deleterious effects of the identified variants, which carry the potential to understand the genotype-phenotype relationships in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Syeda Iqra Hussain
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Nazif Muhammad
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Salah Ud Din Shah
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Fardous Fardous
- Department of Medical Lab Technology, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Sher Alam Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Niamatullah Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Adil U Rehman
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Mehwish Siddique
- Department of Zoology, Government Post Graduate College for Women, Satellite Town, Gujranwala, Pakistan
| | - Shoukat Ali Wasan
- Department of Botany, Faculty of Natural Sciences, Shah Abdul Latif University, Khairpur, Sindh, Pakistan
| | - Rooh Niaz
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Hafiz Ullah
- Gomal Center of Biochemistry and Biotechnology (GCBB), Gomal University D. I. Khan, D. I. Khan, Pakistan
| | - Niamat Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Noor Muhammad
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Usman Mirza
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, N9B 1C4, Canada
| | - Naveed Wasif
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, 89081, Ulm, Germany.
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| | - Saadullah Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Kohat, Khyber Pakhtunkhwa, Pakistan.
| |
Collapse
|
4
|
Tobin JD, Robinson CN, Luttrell-Williams ES, Landry GM, McMartin KE. Lack of efflux of diglycolic acid from proximal tubule cells leads to its accumulation and to toxicity of diethylene glycol. Toxicol Lett 2023; 379:48-55. [PMID: 36958672 DOI: 10.1016/j.toxlet.2023.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
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.
Collapse
Affiliation(s)
- Julie D Tobin
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71130
| | - Corie N Robinson
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71130
| | - Elliot S Luttrell-Williams
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71130
| | - Greg M Landry
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71130
| | - Kenneth E McMartin
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71130.
| |
Collapse
|
5
|
A Novel and Cross-Species Active Mammalian INDY (NaCT) Inhibitor Ameliorates Hepatic Steatosis in Mice with Diet-Induced Obesity. Metabolites 2022; 12:metabo12080732. [PMID: 36005604 PMCID: PMC9413491 DOI: 10.3390/metabo12080732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
Mammalian INDY (mINDY, NaCT, gene symbol SLC13A5) is a potential target for the treatment of metabolically associated fatty liver disease (MAFLD). This study evaluated the effects of a selective, cross-species active, non-competitive, non-substrate-like inhibitor of NaCT. First, the small molecule inhibitor ETG-5773 was evaluated for citrate and succinate uptake and fatty acid synthesis in cell lines expressing both human NaCT and mouse Nact. Once its suitability was established, the inhibitor was evaluated in a diet-induced obesity (DIO) mouse model. DIO mice treated with 15 mg/kg compound ETG-5773 twice daily for 28 days had reduced body weight, fasting blood glucose, and insulin, and improved glucose tolerance. Liver triglycerides were significantly reduced, and body composition was improved by reducing fat mass, supported by a significant reduction in the expression of genes for lipogenesis such as SREBF1 and SCD1. Most of these effects were also evident after a seven-day treatment with the same dose. Further mechanistic investigation in the seven-day study showed increased plasma β-hydroxybutyrate and activated hepatic adenosine monophosphate-activated protein kinase (AMPK), reflecting findings from Indy (−/−) knockout mice. These results suggest that the inhibitor ETG-5773 blocked citrate uptake mediated by mouse and human NaCT to reduce liver steatosis and body fat and improve glucose regulation, proving the concept of NaCT inhibition as a future liver treatment for MAFLD.
Collapse
|
6
|
Thermostability-based binding assays reveal complex interplay of cation, substrate and lipid binding in the bacterial DASS transporter, VcINDY. Biochem J 2021; 478:3847-3867. [PMID: 34643224 PMCID: PMC8652582 DOI: 10.1042/bcj20210061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/04/2022]
Abstract
The divalent anion sodium symporter (DASS) family of transporters (SLC13 family in humans) are key regulators of metabolic homeostasis, disruption of which results in protection from diabetes and obesity, and inhibition of liver cancer cell proliferation. Thus, DASS transporter inhibitors are attractive targets in the treatment of chronic, age-related metabolic diseases. The characterisation of several DASS transporters has revealed variation in the substrate selectivity and flexibility in the coupling ion used to power transport. Here, using the model DASS co-transporter, VcINDY from Vibrio cholerae, we have examined the interplay of the three major interactions that occur during transport: the coupling ion, the substrate, and the lipid environment. Using a series of high-throughput thermostability-based interaction assays, we have shown that substrate binding is Na+-dependent; a requirement that is orchestrated through a combination of electrostatic attraction and Na+-induced priming of the binding site architecture. We have identified novel DASS ligands and revealed that ligand binding is dominated by the requirement of two carboxylate groups in the ligand that are precisely distanced to satisfy carboxylate interaction regions of the substrate-binding site. We have also identified a complex relationship between substrate and lipid interactions, which suggests a dynamic, regulatory role for lipids in VcINDY's transport cycle.
Collapse
|
7
|
Signals from the Circle: Tricarboxylic Acid Cycle Intermediates as Myometabokines. Metabolites 2021; 11:metabo11080474. [PMID: 34436415 PMCID: PMC8398969 DOI: 10.3390/metabo11080474] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022] Open
Abstract
Regular physical activity is an effective strategy to prevent and ameliorate aging-associated diseases. In particular, training increases muscle performance and improves whole-body metabolism. Since exercise affects the whole organism, it has countless health benefits. The systemic effects of exercise can, in part, be explained by communication between the contracting skeletal muscle and other organs and cell types. While small proteins and peptides known as myokines are the most prominent candidates to mediate this tissue cross-talk, recent investigations have paid increasing attention to metabolites. The purpose of this review is to highlight the potential role of tricarboxylic acid (TCA) metabolites as humoral mediators of exercise adaptation processes. We focus on TCA metabolites that are released from human skeletal muscle in response to exercise and provide an overview of their potential auto-, para- or endocrine health-promoting effects.
Collapse
|
8
|
Kang Q, Yang L, Liao H, Yang S, Yang H, Ning Z, Liao C, Wu L. Case Report: Compound Heterozygous Variants of SLC13A3 Identified in a Chinese Patient With Acute Reversible Leukoencephalopathy and α-Ketoglutarate Accumulation. Front Pediatr 2021; 9:801719. [PMID: 34966709 PMCID: PMC8710692 DOI: 10.3389/fped.2021.801719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/22/2021] [Indexed: 11/23/2022] Open
Abstract
Background: SLC13A3 gene encodes the Na+/dicarboxylate cotransporter 3 (NaDC3), which locates on the plasma membrane and is mainly expressed in kidney, astrocytes and the choroid plexus. It imports four to six carbon dicarboxylates together with three Na+ ions into the cytosol. Nowadays, pathogenic variants of SLC13A3 gene were found to cause acute reversible leukoencephalopathy and α-ketoglutarate accumulation (ARLIAK) in patients. Here, we report two novel SLC13A3 variants c.185C>T (p.T62M) and c.331C>T (p.R111*) identified in a Chinese patient with ARLIAK. Case Presentation: The patient was a Chinese girl aged 13 years and 7 months old, who had acute, recurrent neurological deterioration during two febrile episodes. She presented with reversible leukoencephalopathy and increased urinary excretion of α-ketoglutarate. Genetic studies revealed compound heterozygous variants (c.185C>T, p.T62M, and c.331C>T, p.R111*) in SLC13A3, which had not been reported previously. Conclusions: These findings expand the variant spectrum of SLC13A3, providing the basis for the further study of this rare disease.
Collapse
Affiliation(s)
- Qingyun Kang
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Liming Yang
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Hongmei Liao
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Sai Yang
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Haiyang Yang
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Zeshu Ning
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Caishi Liao
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Liwen Wu
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| |
Collapse
|
9
|
Vriend J, Hoogstraten CA, Venrooij KR, van den Berge BT, Govers LP, van Rooij A, Huigen MCDG, Schirris TJJ, Russel FGM, Masereeuw R, Wilmer MJ. Organic anion transporters 1 and 3 influence cellular energy metabolism in renal proximal tubule cells. Biol Chem 2020; 400:1347-1358. [PMID: 30653465 DOI: 10.1515/hsz-2018-0446] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 12/29/2018] [Indexed: 12/19/2022]
Abstract
Organic anion transporters (OATs) 1 and 3 are, besides being uptake transporters, key in several cellular metabolic pathways. The underlying mechanisms are largely unknown. Hence, we used human conditionally immortalized proximal tubule epithelial cells (ciPTEC) overexpressing OAT1 or OAT3 to gain insight into these mechanisms. In ciPTEC-OAT1 and -OAT3, extracellular lactate levels were decreased (by 77% and 71%, respectively), while intracellular ATP levels remained unchanged, suggesting a shift towards an oxidative phenotype upon OAT1 or OAT3 overexpression. This was confirmed by increased respiration of ciPTEC-OAT1 and -OAT3 (1.4-fold), a decreased sensitivity to respiratory inhibition, and characterized by a higher demand on mitochondrial oxidative capacity. In-depth profiling of tricarboxylic acid (TCA) cycle metabolites revealed reduced levels of intermediates converging into α-ketoglutarate in ciPTEC-OAT1 and -OAT3, which via 2-hydroxyglutarate metabolism explains the increased respiration. These interactions with TCA cycle metabolites were in agreement with metabolomic network modeling studies published earlier. Further studies using OAT or oxidative phosphorylation (OXPHOS) inhibitors confirmed our idea that OATs are responsible for increased use and synthesis of α-ketoglutarate. In conclusion, our results indicate an increased α-ketoglutarate efflux by OAT1 and OAT3, resulting in a metabolic shift towards an oxidative phenotype.
Collapse
Affiliation(s)
- Jelle Vriend
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| | - Charlotte A Hoogstraten
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands.,Centre for Systems Biology and Bioenergetics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| | - Kevin R Venrooij
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| | - Bartholomeus T van den Berge
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| | - Larissa P Govers
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| | - Arno van Rooij
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| | - Marleen C D G Huigen
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| | - Tom J J Schirris
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands.,Centre for Systems Biology and Bioenergetics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands.,Centre for Systems Biology and Bioenergetics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Universiteitsweg 99, NL-3584CG, Utrecht, The Netherlands
| | - Martijn J Wilmer
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, NL-6500HB, Nijmegen, The Netherlands
| |
Collapse
|
10
|
Allen SP, Hall B, Woof R, Francis L, Gatto N, Shaw AC, Myszczynska M, Hemingway J, Coldicott I, Willcock A, Job L, Hughes RM, Boschian C, Bayatti N, Heath PR, Bandmann O, Mortiboys H, Ferraiuolo L, Shaw PJ. C9orf72 expansion within astrocytes reduces metabolic flexibility in amyotrophic lateral sclerosis. Brain 2019; 142:3771-3790. [PMID: 31647549 PMCID: PMC6906594 DOI: 10.1093/brain/awz302] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 07/25/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022] Open
Abstract
It is important to understand how the disease process affects the metabolic pathways in amyotrophic lateral sclerosis and whether these pathways can be manipulated to ameliorate disease progression. To analyse the basis of the metabolic defect in amyotrophic lateral sclerosis we used a phenotypic metabolic profiling approach. Using fibroblasts and reprogrammed induced astrocytes from C9orf72 and sporadic amyotrophic lateral sclerosis cases we measured the production rate of reduced nicotinamide adenine dinucleotides (NADH) from 91 potential energy substrates simultaneously. Our screening approach identified that C9orf72 and sporadic amyotrophic lateral sclerosis induced astrocytes have distinct metabolic profiles compared to controls and displayed a loss of metabolic flexibility that was not observed in fibroblast models. This loss of metabolic flexibility, involving defects in adenosine, fructose and glycogen metabolism, as well as disruptions in the membrane transport of mitochondrial specific energy substrates, contributed to increased starvation induced toxicity in C9orf72 induced astrocytes. A reduction in glycogen metabolism was attributed to loss of glycogen phosphorylase and phosphoglucomutase at the protein level in both C9orf72 induced astrocytes and induced neurons. In addition, we found alterations in the levels of fructose metabolism enzymes and a reduction in the methylglyoxal removal enzyme GLO1 in both C9orf72 and sporadic models of disease. Our data show that metabolic flexibility is important in the CNS in times of bioenergetic stress.
Collapse
Affiliation(s)
- Scott P Allen
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Benjamin Hall
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Ryan Woof
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Laura Francis
- The Living Systems Institute, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Noemi Gatto
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Allan C Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Monika Myszczynska
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Jordan Hemingway
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Ian Coldicott
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Amelia Willcock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Lucy Job
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Rachel M Hughes
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Camilla Boschian
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Nadhim Bayatti
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Oliver Bandmann
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield S10 2HQ, UK
| |
Collapse
|
11
|
Dewulf JP, Wiame E, Dorboz I, Elmaleh-Bergès M, Imbard A, Dumitriu D, Rak M, Bourillon A, Helaers R, Malla A, Renaldo F, Boespflug-Tanguy O, Vincent MF, Benoist JF, Wevers RA, Schlessinger A, Van Schaftingen E, Nassogne MC, Schiff M. SLC13A3 variants cause acute reversible leukoencephalopathy and α-ketoglutarate accumulation. Ann Neurol 2019; 85:385-395. [PMID: 30635937 DOI: 10.1002/ana.25412] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 01/22/2023]
Abstract
OBJECTIVE SLC13A3 encodes the plasma membrane Na+ /dicarboxylate cotransporter 3, which imports inside the cell 4 to 6 carbon dicarboxylates as well as N-acetylaspartate (NAA). SLC13A3 is mainly expressed in kidney, in astrocytes, and in the choroid plexus. We describe two unrelated patients presenting with acute, reversible (and recurrent in one) neurological deterioration during a febrile illness. Both patients exhibited a reversible leukoencephalopathy and a urinary excretion of α-ketoglutarate (αKG) that was markedly increased and persisted over time. In one patient, increased concentrations of cerebrospinal fluid NAA and dicarboxylates (including αKG) were observed. Extensive workup was unsuccessful, and a genetic cause was suspected. METHODS Whole exome sequencing (WES) was performed. Our teams were connected through GeneMatcher. RESULTS WES analysis revealed variants in SLC13A3. A homozygous missense mutation (p.Ala254Asp) was found in the first patient. The second patient was heterozygous for another missense mutation (p.Gly548Ser) and an intronic mutation affecting splicing as demonstrated by reverse transcriptase polymerase chain reaction performed in muscle tissue (c.1016 + 3A > G). Mutations and segregation were confirmed by Sanger sequencing. Functional studies performed on HEK293T cells transiently transfected with wild-type and mutant SLC13A3 indicated that the missense mutations caused a marked reduction in the capacity to transport αKG, succinate, and NAA. INTERPRETATION SLC13A3 deficiency causes acute and reversible leukoencephalopathy with marked accumulation of αKG. Urine organic acids (especially αKG and NAA) and SLC13A3 mutations should be screened in patients presenting with unexplained reversible leukoencephalopathy, for which SLC13A3 deficiency is a novel differential diagnosis. ANN NEUROL 2019;85:385-395.
Collapse
Affiliation(s)
- Joseph P Dewulf
- Laboratory of Physiological Chemistry, de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium.,Department of Laboratory Medicine, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Elsa Wiame
- Laboratory of Physiological Chemistry, de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium
| | - Imen Dorboz
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Monique Elmaleh-Bergès
- Department of Pediatric Imaging, Robert Debré University Hospital, Public APHP, Paris, France
| | - Apolline Imbard
- Laboratory of Biochemistry, Robert Debré University Hospital, APHP, France.,Paris-Sud University, Châtenay-Malabry, France
| | - Dana Dumitriu
- Department of Pediatric Imaging, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Malgorzata Rak
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Agnès Bourillon
- Laboratory of Biochemistry, Robert Debré University Hospital, APHP, France.,Paris-Sud University, Châtenay-Malabry, France
| | - Raphaël Helaers
- Human Molecular Genetics, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Alisha Malla
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Florence Renaldo
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,Department of Pediatric Neurology and Metabolic Diseases, Robert Debré University Hospital, APHP, Paris, France.,Reference Center for Leukodystrophies and Rare Leukoencephalopathies, LEUKOFRANCE, Robert Debré University Hospital, APHP, Paris, France
| | - Odile Boespflug-Tanguy
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,Department of Pediatric Neurology and Metabolic Diseases, Robert Debré University Hospital, APHP, Paris, France.,Reference Center for Leukodystrophies and Rare Leukoencephalopathies, LEUKOFRANCE, Robert Debré University Hospital, APHP, Paris, France
| | - Marie-Françoise Vincent
- Department of Laboratory Medicine, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Jean-François Benoist
- Laboratory of Biochemistry, Robert Debré University Hospital, APHP, France.,Paris-Sud University, Châtenay-Malabry, France
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emile Van Schaftingen
- Laboratory of Physiological Chemistry, de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium
| | - Marie-Cécile Nassogne
- Pediatric Neurology Unit, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Manuel Schiff
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,Department of Pediatric Neurology and Metabolic Diseases, Robert Debré University Hospital, APHP, Paris, France.,Reference Center for Inborn Errors of Metabolism, Robert Debré University Hospital, APHP, Paris, France
| |
Collapse
|
12
|
Nieskens TTG, Peters JGP, Dabaghie D, Korte D, Jansen K, Van Asbeck AH, Tavraz NN, Friedrich T, Russel FGM, Masereeuw R, Wilmer MJ. Expression of Organic Anion Transporter 1 or 3 in Human Kidney Proximal Tubule Cells Reduces Cisplatin Sensitivity. Drug Metab Dispos 2018. [PMID: 29514829 DOI: 10.1124/dmd.117.079384] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cisplatin is a cytostatic drug used for treatment of solid organ tumors. The main adverse effect is organic cation transporter 2 (OCT2)-mediated nephrotoxicity, observed in 30% of patients. The contribution of other renal drug transporters is elusive. Here, cisplatin-induced toxicity was evaluated in human-derived conditionally immortalized proximal tubule epithelial cells (ciPTEC) expressing renal drug transporters, including OCT2 and organic anion transporters 1 (OAT1) or 3 (OAT3). Parent ciPTEC demonstrated OCT2-dependent cisplatin toxicity (TC50 34 ± 1 μM after 24-hour exposure), as determined by cell viability. Overexpression of OAT1 and OAT3 resulted in reduced sensitivity to cisplatin (TC50 45 ± 6 and 64 ± 11 μM after 24-hour exposure, respectively). This effect was independent of OAT-mediated transport, as the OAT substrates probenecid and diclofenac did not influence cytotoxicity. Decreased cisplatin sensitivity in OAT-expressing cells was associated directly with a trend toward reduced intracellular cisplatin accumulation, explained by reduced OCT2 gene expression and activity. This was evaluated by Vmax of the OCT2-model substrate ASP+ (23.5 ± 0.1, 13.1 ± 0.3, and 21.6 ± 0.6 minutes-1 in ciPTEC-parent, ciPTEC-OAT1, and ciPTEC-OAT3, respectively). Although gene expression of cisplatin efflux transporter multidrug and toxin extrusion 1 (MATE1) was 16.2 ± 0.3-fold upregulated in ciPTEC-OAT1 and 6.1 ± 0.7-fold in ciPTEC-OAT3, toxicity was unaffected by the MATE substrate pyrimethamine, suggesting that MATE1 does not play a role in the current experimental set-up. In conclusion, OAT expression results in reduced cisplatin sensitivity in renal proximal tubule cells, explained by reduced OCT2-mediated uptake capacity. In vitro drug-induced toxicity studies should consider models that express both OCT and OAT drug transporters.
Collapse
Affiliation(s)
- Tom T G Nieskens
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Janny G P Peters
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Dina Dabaghie
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Daphne Korte
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Katja Jansen
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Alexander H Van Asbeck
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Neslihan N Tavraz
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Thomas Friedrich
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Rosalinde Masereeuw
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Martijn J Wilmer
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| |
Collapse
|
13
|
Uwai Y, Kawasaki T, Nabekura T. D-Malate decreases renal content of α-ketoglutarate, a driving force of organic anion transporters OAT1 and OAT3, resulting in inhibited tubular secretion of phenolsulfonphthalein, in rats. Biopharm Drug Dispos 2017; 38:479-485. [PMID: 28744858 DOI: 10.1002/bdd.2089] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/29/2017] [Accepted: 07/17/2017] [Indexed: 01/20/2023]
Abstract
d-Malate inhibits a Krebs cycle enzyme and the tubular transport of α-ketoglutarate, an intermediate of the Krebs cycle and the driving force for rat organic anion transporter 1 (rOAT1) and rOAT3 in the kidney. This study examined the effects of d-malate on the rat organic anion transport system. The uptake of 6-carboxyfluorescein by HEK293 cells expressing rOAT1 or rOAT3 was not affected by d-malate and l-malate. Up to 60 min after the intravenous injection of phenolsulfonphthalein (PSP), a typical substrate of the renal organic anion transporters, as a bolus to rats, 47.1% of the dose was recovered in the urine, and its renal clearance was estimated to be 8.60 ml/min/kg. d-Malate but not l-malate interfered with its renal excretion, resulting in the delayed elimination of PSP from plasma. No effect of d-malate was recognized on creatinine clearance or the expression level of rOAT3 in the kidney cortex. d-Malate increased the plasma concentration of α-ketoglutarate. In addition, the compound greatly stimulated the renal excretion of α-ketoglutarate, implying that d-malate inhibited its reabsorption. The content of α-ketoglutarate was significantly decreased in the kidney cortex of rats administered d-malate. Collectively, this study shows that d-malate abrogates the tubular secretion of PSP, and the reduction of the renal content of α-ketoglutarate was proposed to be one of the mechanisms. A relationship between the reabsorption of α-ketoglutarate and the basolateral uptake of organic anion in the kidney is suggested.
Collapse
Affiliation(s)
- Yuichi Uwai
- School of Pharmacy, Aichi Gakuin University, 1-100, Kusumoto, Chikusa, Nagoya, 464-8650, Japan
| | - Tatsuya Kawasaki
- School of Pharmacy, Aichi Gakuin University, 1-100, Kusumoto, Chikusa, Nagoya, 464-8650, Japan
| | - Tomohiro Nabekura
- School of Pharmacy, Aichi Gakuin University, 1-100, Kusumoto, Chikusa, Nagoya, 464-8650, Japan
| |
Collapse
|
14
|
Burckhardt BC, Henjakovic M, Hagos Y, Burckhardt G. Differential Interaction of Dantrolene, Glafenine, Nalidixic Acid, and Prazosin with Human Organic Anion Transporters 1 and 3. J Pharmacol Exp Ther 2017. [PMID: 28630284 DOI: 10.1124/jpet.117.241406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In renal proximal tubule cells, the organic anion transporters 1 and 3 (OAT1 and OAT3) in the basolateral membrane and the multidrug resistance-associated protein 4 (MRP4) in the apical membrane share substrates and co-operate in renal drug secretion. We hypothesized that recently identified MRP4 inhibitors dantrolene, glafenine, nalidixic acid, and prazosin also interact with human OAT1 and/or OAT3 stably transfected in human embryonic kidney 293 cells. These four drugs were tested as possible inhibitors of p-[3H]aminohippurate (PAH) and [14C]glutarate uptake by OAT1, and of [3H]estrone-3-sulfate (ES) uptake by OAT3. In addition, we explored whether these drugs decrease the equilibrium distribution of radiolabeled PAH, glutarate, or ES, an approach intended to indirectly suggest drug/substrate exchange through OAT1 and OAT3. With OAT3, a dose-dependent inhibition of [3H]ES uptake and a downward shift in [3H]ES equilibrium were observed, indicating that all four drugs bind to OAT3 and may possibly be translocated. In contrast, the interaction with OAT1 was more complex. With [14C]glutarate as substrate, all four drugs inhibited uptake but only glafenine and nalidixic acid shifted glutarate equilibrium. Using [3H]PAH as a substrate of OAT1, nalidixic acid inhibited but dantrolene, glafenine, and prazosin stimulated uptake. Nalidixic acid decreased equilibrium content of [3H]PAH, suggesting that it may possibly be exchanged by OAT1. Taken together, OAT1 and OAT3 interact with the MRP4 inhibitors dantrolene, glafenine, nalidixic acid, and prazosin, indicating overlapping specificities. At OAT1, more than one binding site must be assumed to explain substrate and drug-dependent stimulation and inhibition of transport activity.
Collapse
Affiliation(s)
- Birgitta C Burckhardt
- Center of Physiology and Pathophysiology, University Medical Center Goettingen, Goettingen, Germany (B.C.B, M.H., Y.H., G.B.); Department I of Internal Medicine, University Medical Center Cologne, Cologne, Germany (M.H.); and PortaCellTec Biosciences GmbH, Goettingen, Germany (Y.H.)
| | - Maja Henjakovic
- Center of Physiology and Pathophysiology, University Medical Center Goettingen, Goettingen, Germany (B.C.B, M.H., Y.H., G.B.); Department I of Internal Medicine, University Medical Center Cologne, Cologne, Germany (M.H.); and PortaCellTec Biosciences GmbH, Goettingen, Germany (Y.H.)
| | - Yohannes Hagos
- Center of Physiology and Pathophysiology, University Medical Center Goettingen, Goettingen, Germany (B.C.B, M.H., Y.H., G.B.); Department I of Internal Medicine, University Medical Center Cologne, Cologne, Germany (M.H.); and PortaCellTec Biosciences GmbH, Goettingen, Germany (Y.H.)
| | - Gerhard Burckhardt
- Center of Physiology and Pathophysiology, University Medical Center Goettingen, Goettingen, Germany (B.C.B, M.H., Y.H., G.B.); Department I of Internal Medicine, University Medical Center Cologne, Cologne, Germany (M.H.); and PortaCellTec Biosciences GmbH, Goettingen, Germany (Y.H.)
| |
Collapse
|
15
|
Burckhardt BC, Henjakovic M, Hagos Y, Burckhardt G. Counter-flow suggests transport of dantrolene and 5-OH dantrolene by the organic anion transporters 2 (OAT2) and 3 (OAT3). Pflugers Arch 2016; 468:1909-1918. [DOI: 10.1007/s00424-016-1894-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/27/2016] [Accepted: 10/11/2016] [Indexed: 11/29/2022]
|
16
|
Klotz J, Porter BE, Colas C, Schlessinger A, Pajor AM. Mutations in the Na(+)/citrate cotransporter NaCT (SLC13A5) in pediatric patients with epilepsy and developmental delay. Mol Med 2016; 22:molmed.2016.00077. [PMID: 27261973 DOI: 10.2119/molmed.2016.00077] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/23/2016] [Indexed: 01/13/2023] Open
Abstract
Mutations in the SLC13A5 gene that codes for the Na(+)/citrate cotransporter, NaCT, are associated with early onset epilepsy, developmental delay and tooth dysplasia in children. In the present study we identify additional SLC13A5 mutations in nine epilepsy patients from six families. To better characterize the syndrome, families with affected children answered questions about the scope of illness and treatment strategies. There are currently no effective treatments, but some anti-epileptic drugs targeting the GABA system reduce seizure frequency. Acetazolamide, a carbonic anhydrase inhibitor and atypical anti-seizure medication decreases seizures in 4 patients. In contrast to previous reports, the ketogenic diet and fasting produce worsening of symptoms. The effects of the mutations on NaCT transport function and protein expression were examined by transient transfections of COS-7 cells. There was no transport activity from any of the mutant transporters, although some of the mutant transporter proteins were present on the plasma membrane. The structural model of NaCT suggests that these mutations can affect helix packing or substrate binding. We tested various treatments, including chemical chaperones and low temperatures, but none improve transport function in the NaCT mutants. Interestingly, coexpression of NaCT and the mutants results in decreased protein expression and activity of the wild-type transporter, indicating functional interaction. In conclusion, our study has identified additional SLC13A5 mutations in patients with chronic epilepsy starting in the neonatal period, with the mutations producing inactive Na(+)/citrate transporters.
Collapse
Affiliation(s)
- Jenna Klotz
- Department of Neurology, Stanford University School of Medicine, Palo Alto, CA 94305
| | - Brenda E Porter
- Department of Neurology, Stanford University School of Medicine, Palo Alto, CA 94305
| | - Claire Colas
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Avner Schlessinger
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ana M Pajor
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla, CA 92130-0718
| |
Collapse
|
17
|
Breljak D, Ljubojević M, Hagos Y, Micek V, Balen Eror D, Vrhovac Madunić I, Brzica H, Karaica D, Radović N, Kraus O, Anzai N, Koepsell H, Burckhardt G, Burckhardt BC, Sabolić I. Distribution of organic anion transporters NaDC3 and OAT1-3 along the human nephron. Am J Physiol Renal Physiol 2016; 311:F227-38. [PMID: 27053689 DOI: 10.1152/ajprenal.00113.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/30/2016] [Indexed: 01/13/2023] Open
Abstract
The initial step in renal secretion of organic anions (OAs) is mediated by transporters in the basolateral membrane (BLM). Contributors to this process are primary active Na(+)-K(+)-ATPase (EC 3.6.3.9), secondary active Na(+)-dicarboxylate cotransporter 3 (NaDC3/SLC13A3), and tertiary active OA transporters (OATs) OAT1/SLC22A6, OAT2/SLC22A7, and OAT3/SLC22A8. In human kidneys, we analyzed the localization of these transporters by immunochemical methods in tissue cryosections and isolated membranes. The specificity of antibodies was validated with human embryonic kidney-293 cells stably transfected with functional OATs. Na(+)-K(+)-ATPase was immunolocalized to the BLM along the entire human nephron. NaDC3-related immunostaining was detected in the BLM of proximal tubules and in the BLM and/or luminal membrane of principal cells in connecting segments and collecting ducts. The thin and thick ascending limbs, macula densa, and distal tubules exhibited no reactivity with the anti-NaDC3 antibody. OAT1-OAT3-related immunostaining in human kidneys was detected only in the BLM of cortical proximal tubules; all three OATs were stained more intensely in S1/S2 segments compared with S3 segment in medullary rays, whereas the S3 segment in the outer stripe remained unstained. Expression of NaDC3, OAT1, OAT2, and OAT3 proteins exhibited considerable interindividual variability in both male and female kidneys, and sex differences in their expression could not be detected. Our experiments provide a side-by-side comparison of basolateral transporters cooperating in renal OA secretion in the human kidney.
Collapse
Affiliation(s)
- Davorka Breljak
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Marija Ljubojević
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Yohannes Hagos
- Center of Physiology and Pathophysiology, Institute of Systemic Physiology and Pathophysiology, University of Göttingen, Göttingen, Germany
| | - Vedran Micek
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Daniela Balen Eror
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Ivana Vrhovac Madunić
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Hrvoje Brzica
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Dean Karaica
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Nikola Radović
- Department of Urology, Clinical Hospital Dubrava, Zagreb, Croatia
| | - Ognjen Kraus
- University Hospital Sisters of Mercy, Zagreb, Croatia
| | - Naohiko Anzai
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Tochigi, Japan; and
| | - Hermann Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute and Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Gerhard Burckhardt
- Center of Physiology and Pathophysiology, Institute of Systemic Physiology and Pathophysiology, University of Göttingen, Göttingen, Germany
| | - Birgitta C Burckhardt
- Center of Physiology and Pathophysiology, Institute of Systemic Physiology and Pathophysiology, University of Göttingen, Göttingen, Germany
| | - Ivan Sabolić
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia;
| |
Collapse
|
18
|
The Chinese Herb Jianpijiedu Contributes to the Regulation of OATP1B2 and ABCC2 in a Rat Model of Orthotopic Transplantation Liver Cancer Pretreated with Food Restriction and Diarrhea. BIOMED RESEARCH INTERNATIONAL 2015; 2015:752850. [PMID: 26665149 PMCID: PMC4664790 DOI: 10.1155/2015/752850] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 10/23/2015] [Accepted: 11/02/2015] [Indexed: 11/17/2022]
Abstract
Traditional Chinese Medicine Jianpijiedu decoction (JPJD) could improve the general status of liver cancer patients in clinics, especially the symptoms of decreased food intake and diarrhea. In this study, our results showed that the survival rate of the liver cancer with food restriction and diarrhea (FRD-LC) rats was lower than the liver cancer (LC) rats, and the tumor volume of the FRD-LC rats was higher than the LC rats. It was also shown that the high dose of JPJD significantly improved the survival rate, weight, and organ weight when compared with FRD-LC-induced rats. Moreover, JPJD administration upregulated the mRNA and protein levels of ABCC2 and downregulated the mRNA and protein levels of OATP1B2 in liver tissues. However, opposite results were observed in the cancer tissues. In conclusion, the study indicated that the Chinese Medicine JPJD could contribute to the rats with liver cancer which were pretreated with food restriction and diarrhea by regulating the expression of ABCC2 and OATP1B2 in liver tissues and cancer tissues.
Collapse
|
19
|
Henjakovic M, Hagos Y, Krick W, Burckhardt G, Burckhardt BC. Human organic anion transporter 2 is distinct from organic anion transporters 1 and 3 with respect to transport function. Am J Physiol Renal Physiol 2015; 309:F843-51. [DOI: 10.1152/ajprenal.00140.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/15/2015] [Indexed: 02/03/2023] Open
Abstract
Phylogentically, organic anion transporter (OAT)1 and OAT3 are closely related, whereas OAT2 is more distant. Experiments with human embryonic kidney-293 cells stably transfected with human OAT1, OAT2, or OAT3 were performed to compare selected transport properties. Common to OAT1, OAT2, and OAT3 is their ability to transport cGMP. OAT2 interacted with prostaglandins, and cGMP uptake was inhibited by PGE2 and PGF2α with IC50 values of 40.8 and 12.7 μM, respectively. OAT1 (IC50: 23.7 μM), OAT2 (IC50: 9.5 μM), and OAT3 (IC50: 1.6 μM) were potently inhibited by MK571, an established multidrug resistance protein inhibitor. OAT2-mediated cGMP uptake was not inhibited by short-chain monocarboxylates and, as opposed to OAT1 and OAT3, not by dicarboxylates. Consequently, OAT2 showed no cGMP/glutarate exchange. OAT1 and OAT3 exhibited a pH and a Cl− dependence with higher substrate uptake at acidic pH and lower substrate uptake in the absence of Cl−, respectively. Such pH and Cl− dependencies were not observed with OAT2. Depolarization of membrane potential by high K+ concentrations in the presence of the K+ ionophore valinomycin left cGMP uptake unaffected. In addition to cGMP, OAT2 transported urate and glutamate, but cGMP/glutamate exchange could not be demonstrated. These experiments suggest that OAT2-mediated cGMP uptake does not occur via exchange with monocarboxylates, dicarboxylates, and hydroxyl ions. The counter anion for electroneutral cGMP uptake remains to be identified.
Collapse
Affiliation(s)
- Maja Henjakovic
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Yohannes Hagos
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfgang Krick
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerhard Burckhardt
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Birgitta C. Burckhardt
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
20
|
Grimm PR, Lazo-Fernandez Y, Delpire E, Wall SM, Dorsey SG, Weinman EJ, Coleman R, Wade JB, Welling PA. Integrated compensatory network is activated in the absence of NCC phosphorylation. J Clin Invest 2015; 125:2136-50. [PMID: 25893600 DOI: 10.1172/jci78558] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 02/09/2015] [Indexed: 12/11/2022] Open
Abstract
Thiazide diuretics are used to treat hypertension; however, compensatory processes in the kidney can limit antihypertensive responses to this class of drugs. Here, we evaluated compensatory pathways in SPAK kinase-deficient mice, which are unable to activate the thiazide-sensitive sodium chloride cotransporter NCC (encoded by Slc12a3). Global transcriptional profiling, combined with biochemical, cell biological, and physiological phenotyping, identified the gene expression signature of the response and revealed how it establishes an adaptive physiology. Salt reabsorption pathways were created by the coordinate induction of a multigene transport system, involving solute carriers (encoded by Slc26a4, Slc4a8, and Slc4a9), carbonic anhydrase isoforms, and V-type H⁺-ATPase subunits in pendrin-positive intercalated cells (PP-ICs) and ENaC subunits in principal cells (PCs). A distal nephron remodeling process and induction of jagged 1/NOTCH signaling, which expands the cortical connecting tubule with PCs and replaces acid-secreting α-ICs with PP-ICs, were partly responsible for the compensation. Salt reabsorption was also activated by induction of an α-ketoglutarate (α-KG) paracrine signaling system. Coordinate regulation of a multigene α-KG synthesis and transport pathway resulted in α-KG secretion into pro-urine, as the α-KG-activated GPCR (Oxgr1) increased on the PP-IC apical surface, allowing paracrine delivery of α-KG to stimulate salt transport. Identification of the integrated compensatory NaCl reabsorption mechanisms provides insight into thiazide diuretic efficacy.
Collapse
|
21
|
Lombardi G, Corona G, Bellu L, Della Puppa A, Pambuku A, Fiduccia P, Bertorelle R, Gardiman MP, D'Avella D, Toffoli G, Zagonel V. Diagnostic value of plasma and urinary 2-hydroxyglutarate to identify patients with isocitrate dehydrogenase-mutated glioma. Oncologist 2015; 20:562-7. [PMID: 25862748 DOI: 10.1634/theoncologist.2014-0266] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 01/15/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Mutant isocitrate dehydrogenase (IDH) 1/2 enzymes can convert α-ketoglutarate into 2-hydroxyglutarate (2HG). The aim of the present study was to explore whether 2HG in plasma and urine could predict the presence of IDH1/2 mutations in patients with glioma. MATERIALS AND METHODS All patients had histological confirmation of glioma and a recent brain magnetic resonance imaging scan showing the neoplastic lesion. Plasma and urine samples were taken from all patients, and the 2HG concentrations were determined using liquid chromatography tandem mass spectrometry. RESULTS A total of 84 patients were enrolled: 38 with R132H-IDH1 mutated and 46 with wild type. Among the 38 patients with mutant IDH1, 21 had high-grade glioma and 17 had low-grade glioma. Among the 46 patients with IDH1 wild-type glioma, 35 and 11 had high- and low-grade glioma, respectively. In all patients, we analyzed the mean 2HG concentration in the plasma, urine, and plasma/urine ratio (Ratio_2HG). We found a significant difference in the Ratio_2HG between patients with and without an IDH1 mutation (22.2 ± 8.7 vs. 15.6 ± 6.8; p < .0001). The optimal cutoff value for Ratio_2HG to identify IDH1 mutation was 19 (sensitivity, 63%; specificity, 76%; accuracy, 70%). In the patients with high-grade glioma only, the optimal cutoff value was 20 (sensitivity, 76%; specificity, 89%; accuracy, 84%; positive predictive value, 80%; negative predictive value, 86%). In 7 of 7 patients with high-grade glioma, we found a correlation between the Ratio_2HG value and the response to treatment. CONCLUSION Ratio_2HG might be a predictor of the presence of IDH1 mutation. The measurement of 2HG could be useful for disease monitoring and also to assess the treatment effects in these patients.
Collapse
Affiliation(s)
- Giuseppe Lombardi
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Giuseppe Corona
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Luisa Bellu
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Alessandro Della Puppa
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Ardi Pambuku
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Pasquale Fiduccia
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Roberta Bertorelle
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Marina Paola Gardiman
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Domenico D'Avella
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Giuseppe Toffoli
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| | - Vittorina Zagonel
- Department of Clinical and Experimental Oncology, Medical Oncology 1, Clinical Trials and Biostatistics Unit, and Molecular Immunology and Oncology Unit, Veneto Institute of Oncology-IRCCS, Padua, Italy; Experimental and Clinical Pharmacology, National Cancer Institute, Aviano, Italy; Neurosurgery Department and Pathology Department, Neurological Sciences, Padua Hospital, Padua, Italy; Neurosurgery Department, University of Padua, Padua, Italy
| |
Collapse
|
22
|
Schwob E, Hagos Y, Burckhardt G, Burckhardt BC. Transporters involved in renal excretion of N-carbamoylglutamate, an orphan drug to treat inborn n-acetylglutamate synthase deficiency. Am J Physiol Renal Physiol 2014; 307:F1373-9. [PMID: 25354943 DOI: 10.1152/ajprenal.00482.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Inborn defects in N-acetylglutamate (NAG) synthase (NAGS) cause a reduction of NAG, an essential cofactor for the initiation of the urea cycle. As a consequence, blood ammonium concentrations are elevated, leading to severe neurological disorders. The orphan drug N-carbamoylglutamate (NCG; Carbaglu), efficiently overcomes NAGS deficiency. However, not much is known about the transporters involved in the uptake, distribution, and elimination of the divalent organic anion NCG. Organic anion-transporting polypeptides (OATPs) as well as organic anion transporters (OATs) working in cooperation with sodium dicarboxylate cotransporter 3 (NaDC3) accept a wide variety of structurally unrelated drugs. To test for possible interactions with OATPs and OATs, the impact of NCG on these transporters in stably transfected human embryonic kidney-293 cells was measured. The two-electrode voltage-clamp technique was used to monitor NCG-mediated currents in Xenopus laevis oocytes that expressed NaDC3. Neither OATPs nor OAT2 and OAT3 interacted with NCG, but OAT1 transported NCG. In addition, NCG was identified as a high-affinity substrate of NaDC3. Preincubation of OAT4-transfected human embryonic kidney-293 cells with NCG showed an increased uptake of estrone sulfate, the reference substrate of OAT4, indicating efflux of NCG by OAT4. In summary, NaDC3 and, to a lesser extent, OAT1 are likely to be responsible for the uptake of NCG from the blood. Efflux of NCG across the luminal membrane into the tubular lumen probably occurs by OAT4 completing renal secretion of this drug.
Collapse
Affiliation(s)
- Elisabeth Schwob
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Yohannes Hagos
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerhard Burckhardt
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Birgitta C Burckhardt
- Institute of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
23
|
Schlessinger A, Sun NN, Colas C, Pajor AM. Determinants of substrate and cation transport in the human Na+/dicarboxylate cotransporter NaDC3. J Biol Chem 2014; 289:16998-7008. [PMID: 24808185 DOI: 10.1074/jbc.m114.554790] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Metabolic intermediates, such as succinate and citrate, regulate important processes ranging from energy metabolism to fatty acid synthesis. Cytosolic concentrations of these metabolites are controlled, in part, by members of the SLC13 gene family. The molecular mechanism underlying Na(+)-coupled di- and tricarboxylate transport by this family is understood poorly. The human Na(+)/dicarboxylate cotransporter NaDC3 (SLC13A3) is found in various tissues, including the kidney, liver, and brain. In addition to citric acid cycle intermediates such as α-ketoglutarate and succinate, NaDC3 transports other compounds into cells, including N-acetyl aspartate, mercaptosuccinate, and glutathione, in keeping with its dual roles in cell nutrition and detoxification. In this study, we construct a homology structural model of NaDC3 on the basis of the structure of the Vibrio cholerae homolog vcINDY. Our computations are followed by experimental testing of the predicted NaDC3 structure and mode of interaction with various substrates. The results of this study show that the substrate and cation binding domains of NaDC3 are composed of residues in the opposing hairpin loops and unwound portions of adjacent helices. Furthermore, these results provide a possible explanation for the differential substrate specificity among dicarboxylate transporters that underpin their diverse biological roles in metabolism and detoxification. The structural model of NaDC3 provides a framework for understanding substrate selectivity and the Na(+)-coupled anion transport mechanism by the human SLC13 family and other key solute carrier transporters.
Collapse
Affiliation(s)
- Avner Schlessinger
- From the Department of Pharmacology and Systems Therapeutics, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029 and
| | - Nina N Sun
- the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92130-0718
| | - Claire Colas
- From the Department of Pharmacology and Systems Therapeutics, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029 and
| | - Ana M Pajor
- the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92130-0718
| |
Collapse
|
24
|
Ingraham L, Li M, Renfro JL, Parker S, Vapurcuyan A, Hanna I, Pelis RM. A Plasma Concentration of α-Ketoglutarate Influences the Kinetic Interaction of Ligands with Organic Anion Transporter 1. Mol Pharmacol 2014; 86:86-95. [DOI: 10.1124/mol.114.091777] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
25
|
Sodium-coupled dicarboxylate and citrate transporters from the SLC13 family. Pflugers Arch 2013; 466:119-30. [PMID: 24114175 DOI: 10.1007/s00424-013-1369-y] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 09/19/2013] [Accepted: 09/23/2013] [Indexed: 12/30/2022]
Abstract
The SLC13 family in humans and other mammals consists of sodium-coupled transporters for anionic substrates: three transporters for dicarboxylates/citrate and two transporters for sulfate. This review will focus on the di- and tricarboxylate transporters: NaDC1 (SLC13A2), NaDC3 (SLC13A3), and NaCT (SLC13A5). The substrates of these transporters are metabolic intermediates of the citric acid cycle, including citrate, succinate, and α-ketoglutarate, which can exert signaling effects through specific receptors or can affect metabolic enzymes directly. The SLC13 transporters are important for regulating plasma, urinary and tissue levels of these metabolites. NaDC1, primarily found on the apical membranes of renal proximal tubule and small intestinal cells, is involved in regulating urinary levels of citrate and plays a role in kidney stone development. NaDC3 has a wider tissue distribution and high substrate affinity compared with NaDC1. NaDC3 participates in drug and xenobiotic excretion through interactions with organic anion transporters. NaCT is primarily a citrate transporter located in the liver and brain, and its activity may regulate metabolic processes. The recent crystal structure of the Vibrio cholerae homolog, VcINDY, provides a new framework for understanding the mechanism of transport in this family. This review summarizes current knowledge of the structure, function, and regulation of the di- and tricarboxylate transporters of the SLC13 family.
Collapse
|
26
|
Hagos Y, Burckhardt G, Burckhardt BC. Human organic anion transporter OAT1 is not responsible for glutathione transport but mediates transport of glutamate derivatives. Am J Physiol Renal Physiol 2013; 304:F403-9. [DOI: 10.1152/ajprenal.00412.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Due to their clearance function, the kidneys are exposed to high concentrations of oxidants and potentially toxic substances. To maintain cellular integrity, renal cells have to be protected by sufficient concentrations of the antioxidant glutathione (GSH). We tested whether GSH or its precursors are taken up by human organic anion transporters 1 (OAT1) and 3 (OAT3) stably expressed in HEK293 cells. GSH did not inhibit uptake of p-aminohippurate (PAH) or of estrone sulfate (ES) in OAT3-transfected HEK293 cells. In OAT1-transfected cells, GSH reduced the uptake of PAH marginally. Among the GSH constituent amino acids, glutamate, cysteine, and glycine, only glutamate inhibited OAT1, but labeled glutamate was not taken up by a probenecid-inhibitable transport system. Thus OAT1 binds glutamate but is unable to translocate it. The GSH precursor dipeptide, cysteinyl glycine (cysgly), and the glutamate derivative N-acetyl glutamate (NAG), inhibited uptake of PAH when present in the medium and trans-stimulated uptake of PAH from the intracellular side, indicating that they are hitherto unrecognized transported substrates of OAT1. N-acetyl aspartate weakly interacted with OAT1, but aspartate did not. NAG inhibited also OAT3, albeit with much lower affinity compared with OAT1, and glutamate did not interact with OAT3 at all. Taken together, human OAT3 and OAT1 cannot be involved in renal GSH extraction from the blood. However, OAT1 could support intracellular GSH synthesis by taking up cysteinyl glycine.
Collapse
Affiliation(s)
- Yohannes Hagos
- Department of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerhard Burckhardt
- Department of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Birgitta C. Burckhardt
- Department of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
27
|
Bobulescu IA, Moe OW. Renal transport of uric acid: evolving concepts and uncertainties. Adv Chronic Kidney Dis 2012; 19:358-71. [PMID: 23089270 PMCID: PMC3619397 DOI: 10.1053/j.ackd.2012.07.009] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 07/17/2012] [Indexed: 02/07/2023]
Abstract
In addition to its role as a metabolic waste product, uric acid has been proposed to be an important molecule with multiple functions in human physiologic and pathophysiologic processes and may be linked to human diseases beyond nephrolithiasis and gout. Uric acid homeostasis is determined by the balance between production, intestinal secretion, and renal excretion. The kidney is an important regulator of circulating uric acid levels by reabsorbing about 90% of filtered urate and being responsible for 60% to 70% of total body uric acid excretion. Defective renal handling of urate is a frequent pathophysiologic factor underpinning hyperuricemia and gout. Despite tremendous advances over the past decade, the molecular mechanisms of renal urate transport are still incompletely understood. Many transport proteins are candidate participants in urate handling, with URAT1 and GLUT9 being the best characterized to date. Understanding these transporters is increasingly important for the practicing clinician as new research unveils their physiologic characteristics, importance in drug action, and genetic association with uric acid levels in human populations. The future may see the introduction of new drugs that act specifically on individual renal urate transporters for the treatment of hyperuricemia and gout.
Collapse
Affiliation(s)
- Ion Alexandru Bobulescu
- Departments of Internal Medicine and Physiology and the Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-8856, USA.
| | | |
Collapse
|
28
|
Selective stabilization of HIF-1α in renal tubular cells by 2-oxoglutarate analogues. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1595-606. [PMID: 22944601 DOI: 10.1016/j.ajpath.2012.07.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 06/07/2012] [Accepted: 07/06/2012] [Indexed: 11/22/2022]
Abstract
The role of proximal versus distal tubular injury in the pathogenesis of acute kidney injury (AKI) is debatable. Inhibition of prolyl hydroxylases that regulate the degradation of hypoxia-inducible transcription factors (HIFs) is a promising therapeutic approach to optimize energy preservation under hypoxia and has successfully been applied to protect kidney structure and function in AKI models. Presently used prolyl hydroxylase inhibitors are lipophilic 2-oxoglutarate analogues (2OGAs) that are widely taken up in cells of most organs. Given the selective expression of organic anion transporters (OATs) in renal proximal tubular cells, we hypothesized that hydrophilic 2OGAs can specifically target proximal tubular cells. We found that cellular hydrophilic 2OGAs uptake depended on OATs and largely confined to the kidney, where it resulted in activation of HIF target genes only in proximal tubular cells. When applied in ischemia-reperfusion experiments, systemically active 2OGA preserved kidney structure and function, but OAT1-transported 2OGA was not protective, suggesting that HIF stabilization in distal tubular rather than proximal tubular cells and/or nontubular cells mediates protective effects. This study provides proof of concept for selective drug targeting of proximal tubular cells on the basis of specific transporters, gives insights into the role of different nephron segments in AKI pathophysiology, and may offer options for long-term HIF stabilization in proximal tubules without confounding effects of erythropoietin induction in peritubular cells and unwarranted extrarenal effects.
Collapse
|
29
|
Hagos Y, Schley G, Schödel J, Krick W, Burckhardt G, Willam C, Burckhardt BC. α-Ketoglutarate-related inhibitors of HIF prolyl hydroxylases are substrates of renal organic anion transporters 1 (OAT1) and 4 (OAT4). Pflugers Arch 2012; 464:367-74. [DOI: 10.1007/s00424-012-1140-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 06/19/2012] [Accepted: 07/24/2012] [Indexed: 01/21/2023]
|
30
|
Burckhardt G. Drug transport by Organic Anion Transporters (OATs). Pharmacol Ther 2012; 136:106-30. [PMID: 22841915 DOI: 10.1016/j.pharmthera.2012.07.010] [Citation(s) in RCA: 255] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 07/10/2012] [Indexed: 02/08/2023]
Abstract
Common to all so far functionally characterized Organic Anion Transporters (OATs) is their broad substrate specificity and their ability to exchange extracellular against intracellular organic anions. Many OATs occur in renal proximal tubules, the site of active drug secretion. Exceptions are murine Oat6 (nasal epithelium), human OAT7 (liver), and rat Oat8 (renal collecting ducts). In human kidneys, OAT1, OAT2, and OAT3 are localized in the basolateral membrane, and OAT4, OAT10, and URAT1 in the apical cell membrane of proximal tubule cells, respectively. In rats and mice, Oat1 and Oat3 are located basolaterally, and Oat2, Oat5, Oat9, Oat10, and Urat1 apically. Several classes of drugs interact with human OAT1-3, including ACE inhibitors, angiotensin II receptor antagonists, diuretics, HMG CoA reductase inhibitors, β-lactam antibiotics, antineoplastic and antiviral drugs, and uricosuric drugs. For most drugs, interaction was demonstrated in vitro by inhibition of OAT-mediated transport of model substrates; for some drugs, transport by OATs was directly proven. Based on IC₅₀ values reported in the literature, OAT1 and OAT3 show comparable affinities for diuretics, cephalosporins, and nonsteroidal anti-inflammatory drugs whereas OAT2 has a lower affinity to most of these compounds. Drug-drug interactions at OAT1 and OAT3 may retard renal drug secretion and cause untoward effects. OAT4, OAT10, and URAT1 in the apical membrane contribute to proximal tubular urate absorption, and OAT10 to nicotinate absorption. OAT4 is in addition able to release drugs, e.g. diuretics, into the tubule lumen.
Collapse
Affiliation(s)
- Gerhard Burckhardt
- Abteilung Vegetative Physiologie und Pathophysiologie, Zentrum Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany.
| |
Collapse
|