1
|
Banjarnahor S, Scherpinski LA, Keller M, König J, Maas R. Differential uptake of arginine derivatives by the human heteromeric amino acid transporter b 0,+AT-rBAT (SLC7A9-SLC3A1). NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03510-z. [PMID: 39480524 DOI: 10.1007/s00210-024-03510-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024]
Abstract
L-arginine and its (patho-)physiologically active derivatives, L-homoarginine and asymmetric dimethylarginine (ADMA), show significant differences in their renal clearance. The underlying molecular mechanisms remain to be elucidated, but selective tubular transport protein-mediated mechanisms likely play a role. In the present study, we investigate the human heteromeric transporter b0,+AT-rBAT (encoded by the SLC7A9 and SLC3A1 genes) as a potential candidate because it is localized in the luminal membrane of human proximal tubule cells and capable of mediating the cellular uptake of amino acids, including L-arginine. Double-transfected Madin-Darby canine kidney (MDCK) cells stably expressing human b0,+AT-rBAT exhibited significant uptake of L-arginine and L-homoarginine, with apparent Km values of 512.6 and 197.0 μM, respectively. On the contrary, ADMA uptake was not saturated up to 4000 μM, with a transport rate > 5 nmol × mg protein-1 × min-1. With an IC50 value of 115.8 μM, L-arginine inhibited L-homoarginine uptake. Conversely, L-arginine only exhibited a partial inhibitory effect on ADMA uptake. Taken together, our data indicate that b0,+AT-rBAT may contribute to the differential renal handling of L-arginine, L-homoarginine, and ADMA.
Collapse
Affiliation(s)
- Sofna Banjarnahor
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- Research Centre for Pharmaceutical Ingredient and Traditional Medicine, Cibinong Science Center, National Research and Innovation Agency (BRIN), 16911, Cibinong, Jawa Barat, Indonesia
| | - Lorenz A Scherpinski
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Max Keller
- Institute of Pharmacy, Universität Regensburg, 93040, Regensburg, Germany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- FAU NeW Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
- FAU NeW Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
| |
Collapse
|
2
|
Lumpuy-Castillo J, Amador-Martínez I, Díaz-Rojas M, Lorenzo O, Pedraza-Chaverri J, Sánchez-Lozada LG, Aparicio-Trejo OE. Role of mitochondria in reno-cardiac diseases: A study of bioenergetics, biogenesis, and GSH signaling in disease transition. Redox Biol 2024; 76:103340. [PMID: 39250857 PMCID: PMC11407069 DOI: 10.1016/j.redox.2024.103340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are global health burdens with rising prevalence. Their bidirectional relationship with cardiovascular dysfunction, manifesting as cardio-renal syndromes (CRS) types 3 and 4, underscores the interconnectedness and interdependence of these vital organ systems. Both the kidney and the heart are critically reliant on mitochondrial function. This organelle is currently recognized as a hub in signaling pathways, with emphasis on the redox regulation mediated by glutathione (GSH). Mitochondrial dysfunction, including impaired bioenergetics, redox, and biogenesis pathways, are central to the progression of AKI to CKD and the development of CRS type 3 and 4. This review delves into the metabolic reprogramming and mitochondrial redox signaling and biogenesis alterations in AKI, CKD, and CRS. We examine the pathophysiological mechanisms involving GSH redox signaling and the AMP-activated protein kinase (AMPK)-sirtuin (SIRT)1/3-peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α) axis in these conditions. Additionally, we explore the therapeutic potential of GSH synthesis inducers in mitigating these mitochondrial dysfunctions, as well as their effects on inflammation and the progression of CKD and CRS types 3 and 4.
Collapse
Affiliation(s)
- Jairo Lumpuy-Castillo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz-Ciberdem, Medicine Department, Autonomous University, 28040, Madrid, Spain.
| | - Isabel Amador-Martínez
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico; Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04510, Mexico City, Mexico.
| | - Miriam Díaz-Rojas
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 43210, Columbus, Ohio, USA.
| | - Oscar Lorenzo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz-Ciberdem, Medicine Department, Autonomous University, 28040, Madrid, Spain.
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04510, Mexico City, Mexico.
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico.
| | - Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico.
| |
Collapse
|
3
|
Chait AR, Hassig S, Cubillos J, Jain R, Quarrier SO. Cystinuria Complicated by Anuria From Bilateral Obstructing Stones Requiring Bilateral Mini Percutaneous Nephrolithotomy in a 22-Month-Old. Urology 2024; 185:88-90. [PMID: 38281667 DOI: 10.1016/j.urology.2023.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/31/2023] [Indexed: 01/30/2024]
Abstract
Pediatric nephrolithiasis is increasing in incidence and presents differently compared to adults. We report a case of nephrolithiasis in a pediatric patient, presenting with complaints of emesis, anuria, hematuria, and abdominal distension, leading to a diagnosis of bilateral obstructing cystine stones requiring bilateral percutaneous nephrolithotomy. Pediatric patients with anuria should be evaluated for bilateral nephrolithiasis as an etiology. Calculous anuria requires prompt recognition of the pathologic process and relief of the obstruction with close follow-up and supportive care until definitive stone management. Bilateral percutaneous nephrolithotomy can provide definitive surgical intervention without significant morbidity.
Collapse
Affiliation(s)
- Alexander R Chait
- Department of Urology, University of Rochester Medical Center, Rochester, NY.
| | - Stephen Hassig
- Department of Urology, University of Rochester Medical Center, Rochester, NY
| | - Jimena Cubillos
- Department of Urology, University of Rochester Medical Center, Rochester, NY
| | - Rajat Jain
- Department of Urology, University of Rochester Medical Center, Rochester, NY
| | - Scott O Quarrier
- Department of Urology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
4
|
Liu G, Sharma MK, Tompkins YH, Teng PY, Kim WK. Impacts of varying methionine to cysteine supplementation ratios on growth performance, oxidative status, intestinal health, and gene expression of immune response and methionine metabolism in broilers under Eimeria spp. challenge. Poult Sci 2024; 103:103300. [PMID: 38100947 PMCID: PMC10762478 DOI: 10.1016/j.psj.2023.103300] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
A study was conducted to investigate effects of different methionine (Met) to cysteine (Cys) supplementation ratios (MCR) on growth performance, oxidative status, intestinal health, immune responses, and methionine metabolism in broilers under Eimeria challenge. A total of 720 male Cobb500 broilers (14-day-old) were allocated in a 2 × 5 factorial arrangement (5 diets, with or without challenge) with 6 replicates per treatment. The total sulfur amino acid concentrations were consistent across treatments meeting the breeder's recommendation, only MCR varied. The diets were labeled as MET100; MET75; MET50; MET25; and MET0, representing MCR of 100:0; 75:25; 50:50; 25:75; and 0:100, respectively. Data were analyzed by 2-way ANOVA and orthogonal polynomial contrast. Growth performance declined linearly or quadratically as MCR decreased (P < 0.01). On 6-day postinoculation (DPI), interaction effects (P < 0.01) were found; BW and body weight gain were lower in MET0 compared to the other treatments in the nonchallenged groups, whereas not in the challenged groups. On 6 and 9 DPI, serum total antioxidant capacity linearly decreased as MCR decreased (P < 0.05). Hepatic activities of glutathione peroxidase on 6 DPI and superoxide dismutase on 9 DPI changed quadratically as MCR decreased (P < 0.05). The digestibility of Met linearly decreased whereas the digestibility of Cys linearly increased as MCR decreased. The ileal crypt depth linearly decreased as MCR decreased (P < 0.01) on 6 DPI. The expression of transforming growth factor beta on 6 and 9 DPI, tumor necrotic factor alpha and interleukin 10 on 9 DPI changed quadratically as MCR decreased (P < 0.05). Eimeria challenge increased expression of Met adenosyltransferase and cystathionine gamma-lyase, whereas decreasing the expression of other Met metabolism genes (P < 0.01) on 6 DPI. Expression of Met metabolism genes linearly increased as MCR decreased (P < 0.05). In conclusion, different Met to Cys supplementation ratios exerted linearly or quadratically effects on the growth performance, oxidative status, intestinal health, and metabolism of Met in broiler chickens under Eimeria infection.
Collapse
Affiliation(s)
- Guanchen Liu
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Milan K Sharma
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Yuguo H Tompkins
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Po-Yun Teng
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Woo K Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
5
|
Zhang Z, Zheng R, Zhu C, Geng H, Xu G. Lipidomics characterization of the lipid metabolism profiles in a cystinuria rat model: Precalculus damage in the kidney of cystinuria. Prostaglandins Other Lipid Mediat 2022; 162:106651. [PMID: 35680078 DOI: 10.1016/j.prostaglandins.2022.106651] [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: 12/19/2021] [Revised: 05/20/2022] [Accepted: 06/02/2022] [Indexed: 10/18/2022]
Abstract
Cystinuria is a genetic disorder of cystine transport, including defective protein b0,+AT (encoded by SLC7A9), and/or rBAT (encoded by SLC3A1). Patients present hyperexcretion of cystine in the urine, recurrent cystine lithiasis, and progressive decline in kidney function. Moreover, heterodimer transport is defective. To date, little omics data are accessible regarding this metabolic disease caused by membrane proteins. Since membrane function is closely related to changes in the lipidome, we decided to explore the changes in kidney tissue of a self-established cystinuria rat model by performing lipidomic analysis by LC-MS/MS. Our results demonstrated that Slc7a9 deficiency changed the lipid profile of the renal cortex and induced vital modifications in the lipidome, including major alterations in ChE, LPA, and PA. Among those alterations, this lipidomic study highlights the lipid changes that participate in inflammatory responses during cystinuria. As a result, lipid research, perhaps has great potential, for it may lead to the identification of novel therapeutic targets for the prevention and treatment of cystinuria.
Collapse
Affiliation(s)
- Zihan Zhang
- Shanghai Jiaotong University School of Medicine, China
| | - Rui Zheng
- Department of Pediatric Urology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Caihua Zhu
- Shanghai Applied Protein Technology Co., Ltd., 201100, China
| | - Hongquan Geng
- Shanghai Jiaotong University School of Medicine, China; Department of Pediatric Urology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, China.
| | - Guofeng Xu
- Shanghai Jiaotong University School of Medicine, China; Department of Pediatric Urology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, China.
| |
Collapse
|
6
|
Differences in renal cortex transcriptional profiling of wild-type and novel type B cystinuria model rats. Urolithiasis 2022; 50:279-291. [PMID: 35416493 PMCID: PMC9110498 DOI: 10.1007/s00240-022-01321-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 03/03/2022] [Indexed: 11/20/2022]
Abstract
Cystinuria is a genetic disorder of cystine transport that accounts for 1–2% of all cases of renal lithiasis. It is characterized by hyperexcretion of cystine in urine and recurrent cystine lithiasis. Defective transport of cystine into epithelial cells of renal tubules occurs because of mutations of the transport heterodimer, including protein b0,+AT (encoded by SLC7A9) and rBAT (encoded by SLC3A1) linked through a covalent disulfide bond. Study generated a novel type B cystinuria rat model by artificially deleting 7 bp of Slc7a9 gene exon 3 using the CRISPR-Cas9 system, and those Slc7a9-deficient rats were proved to be similar with cystinuria in terms of genome, transcriptome, translation, and biologic phenotypes with no off-target editing. Subsequent comparisons of renal histopathology indicated model rats gained typical secondary changes as medullary fibrosis with no stone formation. A total of 689 DEGs (383 upregulated and 306 downregulated) were differentially expressed in the renal cortex of cystinuria rats. In accordance with the functional annotation of DEGs, the potential role of glutathione metabolism processes in the kidney of cystinuria rat model was proposed, and KEGG analysis results showed that knock-out of Slc7a9 gene triggered more biological changes which has not been studied. In short, for the first time, a rat model and its transcriptional database that mimics the pathogenesis and clinical consequences of human type B cystinuria were generated.
Collapse
|
7
|
Correia MJ, Pimpão AB, Fernandes DGF, Morello J, Sequeira CO, Calado J, Antunes AMM, Almeida MS, Branco P, Monteiro EC, Vicente JB, Serpa J, Pereira SA. Cysteine as a Multifaceted Player in Kidney, the Cysteine-Related Thiolome and Its Implications for Precision Medicine. Molecules 2022; 27:1416. [PMID: 35209204 PMCID: PMC8874463 DOI: 10.3390/molecules27041416] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/16/2022] Open
Abstract
In this review encouraged by original data, we first provided in vivo evidence that the kidney, comparative to the liver or brain, is an organ particularly rich in cysteine. In the kidney, the total availability of cysteine was higher in cortex tissue than in the medulla and distributed in free reduced, free oxidized and protein-bound fractions (in descending order). Next, we provided a comprehensive integrated review on the evidence that supports the reliance on cysteine of the kidney beyond cysteine antioxidant properties, highlighting the relevance of cysteine and its renal metabolism in the control of cysteine excess in the body as a pivotal source of metabolites to kidney biomass and bioenergetics and a promoter of adaptive responses to stressors. This view might translate into novel perspectives on the mechanisms of kidney function and blood pressure regulation and on clinical implications of the cysteine-related thiolome as a tool in precision medicine.
Collapse
Affiliation(s)
- Maria João Correia
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - António B. Pimpão
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Dalila G. F. Fernandes
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), 2780-157 Oeiras, Portugal; (D.G.F.F.); (J.B.V.)
| | - Judit Morello
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Catarina O. Sequeira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Joaquim Calado
- Centre for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, Nova Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal;
- Nephrology Department, Centro Hospitalar Universitário de Lisboa Central, 1069-166 Lisboa, Portugal
| | - Alexandra M. M. Antunes
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, 1049-001 Lisboa, Portugal;
| | - Manuel S. Almeida
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal
| | - Patrícia Branco
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal
| | - Emília C. Monteiro
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - João B. Vicente
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), 2780-157 Oeiras, Portugal; (D.G.F.F.); (J.B.V.)
| | - Jacinta Serpa
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), 1099-023 Lisboa, Portugal
| | - Sofia A. Pereira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| |
Collapse
|
8
|
Parker JL, Deme JC, Kolokouris D, Kuteyi G, Biggin PC, Lea SM, Newstead S. Molecular basis for redox control by the human cystine/glutamate antiporter system xc . Nat Commun 2021; 12:7147. [PMID: 34880232 PMCID: PMC8654953 DOI: 10.1038/s41467-021-27414-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Cysteine plays an essential role in cellular redox homoeostasis as a key constituent of the tripeptide glutathione (GSH). A rate limiting step in cellular GSH synthesis is the availability of cysteine. However, circulating cysteine exists in the blood as the oxidised di-peptide cystine, requiring specialised transport systems for its import into the cell. System xc- is a dedicated cystine transporter, importing cystine in exchange for intracellular glutamate. To counteract elevated levels of reactive oxygen species in cancerous cells system xc- is frequently upregulated, making it an attractive target for anticancer therapies. However, the molecular basis for ligand recognition remains elusive, hampering efforts to specifically target this transport system. Here we present the cryo-EM structure of system xc- in both the apo and glutamate bound states. Structural comparisons reveal an allosteric mechanism for ligand discrimination, supported by molecular dynamics and cell-based assays, establishing a mechanism for cystine transport in human cells.
Collapse
Affiliation(s)
- Joanne L Parker
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK.
| | - Justin C Deme
- Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
- Central Oxford Structural Molecular Imaging Centre, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | | | - Gabriel Kuteyi
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Philip C Biggin
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Susan M Lea
- Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
- Central Oxford Structural Molecular Imaging Centre, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA.
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK.
| |
Collapse
|
9
|
Ellingsen S, Narawane S, Fjose A, Verri T, Rønnestad I. The zebrafish cationic amino acid transporter/glycoprotein-associated family: sequence and spatiotemporal distribution during development of the transport system b 0,+ (slc3a1/slc7a9). FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:1507-1525. [PMID: 34338990 PMCID: PMC8478756 DOI: 10.1007/s10695-021-00984-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 06/29/2021] [Indexed: 06/12/2023]
Abstract
System b0,+ absorbs lysine, arginine, ornithine, and cystine, as well as some (large) neutral amino acids in the mammalian kidney and intestine. It is a heteromeric amino acid transporter made of the heavy subunit SLC3A1/rBAT and the light subunit SLC7A9/b0,+AT. Mutations in these two genes can cause cystinuria in mammals. To extend information on this transport system to teleost fish, we focused on the slc3a1 and slc7a9 genes by performing comparative and phylogenetic sequence analysis, investigating gene conservation during evolution (synteny), and defining early expression patterns during zebrafish (Danio rerio) development. Notably, we found that slc3a1 and slc7a9 are non-duplicated in the zebrafish genome. Whole-mount in situ hybridization detected co-localized expression of slc3a1 and slc7a9 in pronephric ducts at 24 h post-fertilization and in the proximal convoluted tubule at 3 days post-fertilization (dpf). Notably, both the genes showed co-localized expression in epithelial cells in the gut primordium at 3 dpf and in the intestine at 5 dpf (onset of exogenous feeding). Taken together, these results highlight the value of slc3a1 and slc7a9 as markers of zebrafish kidney and intestine development and show promise for establishing new zebrafish tools that can aid in the rapid screening(s) of substrates. Importantly, such studies will help clarify the complex interplay between the absorption of dibasic amino acids, cystine, and (large) neutral amino acids and the effect(s) of such nutrients on organismal growth.
Collapse
Affiliation(s)
- Ståle Ellingsen
- Department of Molecular Biology, University of Bergen, Postbox 7803, N-5020, Bergen, Norway
- Department of Biological Sciences, University of Bergen, Postbox 7803, N-5020, Bergen, Norway
| | - Shailesh Narawane
- Department of Molecular Biology, University of Bergen, Postbox 7803, N-5020, Bergen, Norway
| | - Anders Fjose
- Department of Molecular Biology, University of Bergen, Postbox 7803, N-5020, Bergen, Norway
- Department of Biological Sciences, University of Bergen, Postbox 7803, N-5020, Bergen, Norway
| | - Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Prov.le Lecce-Monteroni, 73100, Lecce, Italy
| | - Ivar Rønnestad
- Department of Biological Sciences, University of Bergen, Postbox 7803, N-5020, Bergen, Norway.
| |
Collapse
|
10
|
Kovaříková S, Maršálek P, Vrbová K. Cystinuria in Dogs and Cats: What Do We Know after Almost 200 Years? Animals (Basel) 2021; 11:2437. [PMID: 34438894 PMCID: PMC8388795 DOI: 10.3390/ani11082437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/08/2021] [Accepted: 08/13/2021] [Indexed: 11/18/2022] Open
Abstract
The purpose of this review is to summarize current knowledge on canine and feline cystinuria from available scientific reports. Cystinuria is an inherited metabolic defect characterized by abnormal intestinal and renal amino acid transport in which cystine and the dibasic amino acids ornithine, lysine, and arginine are involved (COLA). At a normal urine pH, ornithine, lysine, and arginine are soluble, but cysteine forms a dimer, cystine, which is relatively insoluble, resulting in crystal precipitation. Mutations in genes coding COLA transporter and the mode of inheritance were identified only in some canine breeds. Cystinuric dogs may form uroliths (mostly in lower urinary tract) which are associated with typical clinical symptoms. The prevalence of cystine urolithiasis is much higher in European countries (up to 14% according to the recent reports) when compared to North America (United States and Canada) where it is approximately 1-3%. Cystinuria may be diagnosed by the detection of cystine urolithiasis, cystine crystalluria, assessment of amino aciduria, or using genetic tests. The management of cystinuria is aimed at urolith removal or dissolution which may be reached by dietary changes or medical treatment. In dogs with androgen-dependent cystinuria, castration will help. In cats, cystinuria occurs less frequently in comparison with dogs.
Collapse
Affiliation(s)
- Simona Kovaříková
- Department of Animal Protection and Welfare and Veterinary Public Health, Faculty of Veterinary Hygiene and Ecology, University of Veterinary Sciences, 612 42 Brno, Czech Republic;
| | - Petr Maršálek
- Department of Animal Protection and Welfare and Veterinary Public Health, Faculty of Veterinary Hygiene and Ecology, University of Veterinary Sciences, 612 42 Brno, Czech Republic;
| | - Kateřina Vrbová
- Faculty of Veterinary Medicine, University of Veterinary Sciences, 612 42 Brno, Czech Republic;
| |
Collapse
|
11
|
Pizzagalli MD, Bensimon A, Superti‐Furga G. A guide to plasma membrane solute carrier proteins. FEBS J 2021; 288:2784-2835. [PMID: 32810346 PMCID: PMC8246967 DOI: 10.1111/febs.15531] [Citation(s) in RCA: 186] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
This review aims to serve as an introduction to the solute carrier proteins (SLC) superfamily of transporter proteins and their roles in human cells. The SLC superfamily currently includes 458 transport proteins in 65 families that carry a wide variety of substances across cellular membranes. While members of this superfamily are found throughout cellular organelles, this review focuses on transporters expressed at the plasma membrane. At the cell surface, SLC proteins may be viewed as gatekeepers of the cellular milieu, dynamically responding to different metabolic states. With altered metabolism being one of the hallmarks of cancer, we also briefly review the roles that surface SLC proteins play in the development and progression of cancer through their influence on regulating metabolism and environmental conditions.
Collapse
Affiliation(s)
- Mattia D. Pizzagalli
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Ariel Bensimon
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Giulio Superti‐Furga
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Center for Physiology and PharmacologyMedical University of ViennaAustria
| |
Collapse
|
12
|
Prot-Bertoye C, Daudon M, Tostivint I, Dousseaux MP, Defazio J, Traxer O, Knebelmann B, Courbebaisse M. [Cystinuria]. Nephrol Ther 2021; 17S:S100-S107. [PMID: 33910689 DOI: 10.1016/j.nephro.2020.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/01/2020] [Indexed: 10/21/2022]
Abstract
Cystinuria is the most common monogenic nephrolithiasis disorder. Because of its poor solubility at a typical urine pH of less than 7, cystine excretion results in recurrent urinary cystine stone formation. A high prevalence of high blood pressure and of chronic kidney disease has been reported in these patients. Alkaline hyperdiuresis remains the cornerstone of the preventive medical treatment. To reach a urine pH between 7.5 and 8 and a urine specific gravity less than or equal to 1.005 should be the goal of medical treatment. D-penicillamine and tiopronin, two cysteine-binding thiol agents, should be considered as second line treatments with frequent adverse events that should be closely monitored.
Collapse
Affiliation(s)
- Caroline Prot-Bertoye
- Service de physiologie - explorations fonctionnelles rénales et métaboliques, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France.
| | - Michel Daudon
- Service de physiologie-explorations fonctionnelles, hôpital Tenon, 4, rue de la Chine, 75020 Paris, France
| | - Isabelle Tostivint
- Service de néphrologie, hôpital de la Pitié-Salpêtrière, 149, boulevard de l'Hôpital, 75013 Paris, France
| | - Marie-Paule Dousseaux
- Service de néphrologie, hôpital de la Pitié-Salpêtrière, 149, boulevard de l'Hôpital, 75013 Paris, France
| | - Jérôme Defazio
- Association pour l'information et la recherche sur les maladies génétiques (AIRG-France), BP 78, 75261 Paris cedex 06, France
| | - Olivier Traxer
- Service d'urologie, hôpital Tenon, 4, rue de la Chine, 75020 Paris, France
| | - Bertrand Knebelmann
- Service de néphrologie, hôpital Necker, 149, rue de Sèvres, 75015 Paris, France
| | - Marie Courbebaisse
- Service de physiologie - explorations fonctionnelles rénales et métaboliques, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France
| |
Collapse
|
13
|
Fairweather SJ, Shah N, Brӧer S. Heteromeric Solute Carriers: Function, Structure, Pathology and Pharmacology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 21:13-127. [PMID: 33052588 DOI: 10.1007/5584_2020_584] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Solute carriers form one of three major superfamilies of membrane transporters in humans, and include uniporters, exchangers and symporters. Following several decades of molecular characterisation, multiple solute carriers that form obligatory heteromers with unrelated subunits are emerging as a distinctive principle of membrane transporter assembly. Here we comprehensively review experimentally established heteromeric solute carriers: SLC3-SLC7 amino acid exchangers, SLC16 monocarboxylate/H+ symporters and basigin/embigin, SLC4A1 (AE1) and glycophorin A exchanger, SLC51 heteromer Ost α-Ost β uniporter, and SLC6 heteromeric symporters. The review covers the history of the heteromer discovery, transporter physiology, structure, disease associations and pharmacology - all with a focus on the heteromeric assembly. The cellular locations, requirements for complex formation, and the functional role of dimerization are extensively detailed, including analysis of the first complete heteromer structures, the SLC7-SLC3 family transporters LAT1-4F2hc, b0,+AT-rBAT and the SLC6 family heteromer B0AT1-ACE2. We present a systematic analysis of the structural and functional aspects of heteromeric solute carriers and conclude with common principles of their functional roles and structural architecture.
Collapse
Affiliation(s)
- Stephen J Fairweather
- Research School of Biology, Australian National University, Canberra, ACT, Australia. .,Resarch School of Chemistry, Australian National University, Canberra, ACT, Australia.
| | - Nishank Shah
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Stefan Brӧer
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
14
|
Evaluation of Dissolution Profiles of a Newly Developed Solid Oral Immediate-Release Formula Containing Alpha-Lipoic Acid. Processes (Basel) 2021. [DOI: 10.3390/pr9010176] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alpha-lipoic acid (ALA, thioctic acid), a naturally-occurring essential dithiol compound, has become a common ingredient in many pharmaceutical and food supplement products (FSP), used in oxidative stress-dependent pathologies; oral bioavailability of ALA is limited by pharmacokinetic particularities that reduce its therapeutic efficacy-reduced solubility, lack of gastric stability and hepatic degradation, doubled by formulation hinders. The objectives were to develop a solid oral 600 mg ALA FSP to obtain an optimal pharmaceutical profile compared to a reference listed drug (RLD) with a similarity factor f2 50. A comparative dissolution study was performed; an HPLC method was used for ALA quantification. After planning combinatory simulations (formulation stage), two prototype formulas (#1 and #2) were manufactured and further optimized by adjusting ALA physical characteristics and the excipients quantities (#3 and #4) in order to achieve the Quality Target Product Profile. A misshapen of ALA’s in vitro release was observed for #3 Formula (f2 = 31.6); the optimal profile was obtained for Formula #4 (f2 = 58.5). A simple quantitative formula is not enough to assure good ALA bioavailability; the formulation needs multiple compounding modulations under physicochemical compatibility algorithms, with multiple dissolution profiles testing back-ups. It is essential to ensure a formulation with an in vitro dissolution comparable with the RLD, allowing the compound to reach its target level to assure the optimum claimed antioxidant activity of ALA at the cellular level, even for food supplement formulations.
Collapse
|
15
|
Transport of L-Arginine Related Cardiovascular Risk Markers. J Clin Med 2020; 9:jcm9123975. [PMID: 33302555 PMCID: PMC7764698 DOI: 10.3390/jcm9123975] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
L-arginine and its derivatives, asymmetric and symmetric dimethylarginine (ADMA and SDMA) and L-homoarginine, have emerged as cardiovascular biomarkers linked to cardiovascular outcomes and various metabolic and functional pathways such as NO-mediated endothelial function. Cellular uptake and efflux of L-arginine and its derivatives are facilitated by transport proteins. In this respect the cationic amino acid transporters CAT1 and CAT2 (SLC7A1 and SLC7A2) and the system y+L amino acid transporters (SLC7A6 and SLC7A7) have been most extensively investigated, so far, but the number of transporters shown to mediate the transport of L-arginine and its derivatives is constantly increasing. In the present review we assess the growing body of evidence regarding the function, expression, and clinical relevance of these transporters and their possible relation to cardiovascular diseases.
Collapse
|
16
|
Wu KC, Reisman SA, Klaassen CD. Tissue distribution, hormonal regulation, ontogeny, diurnal expression, and induction of mouse cystine transporters Slc3a1 and Slc7a9. Free Radic Res 2020; 54:525-534. [PMID: 32873097 DOI: 10.1080/10715762.2020.1812597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Slc7a11 (xCT) and Slc3a1 (rBAT) are cystine uptake transporters that maintain intracellular concentrations of cysteine, the rate-limiting amino acid in glutathione synthesis. This study was conducted to first determine the tissue distribution of the two transporters in male and female mice. Because Slc3a1 was the primary cystine transporter in liver, its sex-divergent expression, ontogeny, diurnal rhythm and whether its mRNA expression is altered by transcription factors (AhR, CAR, PXR, PPARα, and Nrf2) was also investigated. Slc7a11 was expressed highest in brain and gonads. Slc3a1 was expressed highest in kidney and intestine, followed by liver. Duodenal and hepatic Slc3a1 was higher in females than males. Hepatic Slc3a1 was high during darkness and low during daytime. Hepatic Scl3a1 was lowest pre-birth, increased to near maximal levels at birth, decreased back to pre-birth levels between Days 3-10, and then returned to peak levels by Day 45. Except for CAR, activation of transcription factors did not increase hepatic mRNA expression of Slc3a1. Chemical activation of CAR significantly induced Slc3a1 1.4-fold in wild-type but not CAR-null mice. Slc3a1 mRNA was higher in livers of AhR- and Nrf2-null mice compared to wild-type mice. High doses of diquat but not acetaminophen induced Slc3a1, suggesting Slc3a1 may respond to oxidative stress but not necessarily to GSH depletion. Overall, Slc7a11 is mainly expressed in brain and gonads, whereas Slc3a1 is mainly expressed in kidney, small intestine and liver, and its hepatic expression is regulated by diurnal rhythm and certain xenobiotic treatments.
Collapse
Affiliation(s)
- Kai Connie Wu
- Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, University of Kansas, Kansas City, MO, USA
| | - Scott A Reisman
- Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, University of Kansas, Kansas City, MO, USA
| | - Curtis D Klaassen
- Department of Pharmacology, Toxicology and Therapeutics, School of Medicine, University of Kansas, Kansas City, MO, USA
| |
Collapse
|
17
|
Structural basis for amino acid exchange by a human heteromeric amino acid transporter. Proc Natl Acad Sci U S A 2020; 117:21281-21287. [PMID: 32817565 DOI: 10.1073/pnas.2008111117] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Heteromeric amino acid transporters (HATs) comprise a group of membrane proteins that belong to the solute carrier (SLC) superfamily. They are formed by two different protein components: a light chain subunit from an SLC7 family member and a heavy chain subunit from the SLC3 family. The light chain constitutes the transport subunit whereas the heavy chain mediates trafficking to the plasma membrane and maturation of the functional complex. Mutation, malfunction, and dysregulation of HATs are associated with a wide range of pathologies or represent the direct cause of inherited and acquired disorders. Here we report the cryogenic electron microscopy structure of the neutral and basic amino acid transport complex (b[0,+]AT1-rBAT) which reveals a heterotetrameric protein assembly composed of two heavy and light chain subunits, respectively. The previously uncharacterized interaction between two HAT units is mediated via dimerization of the heavy chain subunits and does not include participation of the light chain subunits. The b(0,+)AT1 transporter adopts a LeuT fold and is captured in an inward-facing conformation. We identify an amino-acid-binding pocket that is formed by transmembrane helices 1, 6, and 10 and conserved among SLC7 transporters.
Collapse
|
18
|
Effects of Ischemia-Reperfusion on Tubular Cell Membrane Transporters and Consequences in Kidney Transplantation. J Clin Med 2020; 9:jcm9082610. [PMID: 32806541 PMCID: PMC7464608 DOI: 10.3390/jcm9082610] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemia-reperfusion (IR)-induced acute kidney injury (IRI) is an inevitable event in kidney transplantation. It is a complex pathophysiological process associated with numerous structural and metabolic changes that have a profound influence on the early and the late function of the transplanted kidney. Proximal tubular cells are particularly sensitive to IRI. These cells are involved in renal and whole-body homeostasis, detoxification processes and drugs elimination by a transporter-dependent, transcellular transport system involving Solute Carriers (SLCs) and ATP Binding Cassettes (ABCs) transporters. Numerous studies conducted mainly in animal models suggested that IRI causes decreased expression and activity of some major tubular transporters. This could favor uremic toxins accumulation and renal metabolic alterations or impact the pharmacokinetic/toxicity of drugs used in transplantation. It is of particular importance to understand the underlying mechanisms and effects of IR on tubular transporters in order to improve the mechanistic understanding of IRI pathophysiology, identify biomarkers of graft function or promote the design and development of novel and effective therapies. Modulation of transporters’ activity could thus be a new therapeutic opportunity to attenuate kidney injury during IR.
Collapse
|
19
|
Fagundes NS, Milfort MC, Williams SM, Da Costa MJ, Fuller AL, Menten JF, Rekaya R, Aggrey SE. Dietary methionine level alters growth, digestibility, and gene expression of amino acid transporters in meat-type chickens. Poult Sci 2020; 99:67-75. [PMID: 32416854 PMCID: PMC7587823 DOI: 10.3382/ps/pez588] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 10/01/2019] [Indexed: 01/16/2023] Open
Abstract
Imbalance in nutrients can affect digestibility of amino acids by altering gene expression of amino acid transporters. We investigated digestibility and molecular transporters of essential amino acids in chickens fed a methionine-deficient diet. A total of 40 chicks (23 D old) were randomly assigned to either a control (0.49% methionine) or a deficient (0.28%) diet until 41 D when they were sampled for Pectoralis (P.) major, kidney, ileum, and hypothalamus for mRNA expression analysis. The ileal content was collected for apparent ileal digestibility (AID) analysis. Birds fed the deficient diet had reduced growth and worse feed efficiency compared to control. The AID of methionine was similar between both groups. The AID of other essential amino acids was higher in the deficient group than control. mRNA expression of b0,+ AT and LAT4 were upregulated in the ileum and kidney but LAT1 was downregulated only in kidney of the deficient group compared to control. In the P. major, SNAT1, SNAT2, and CAT1 were upregulated in the deficient group compared to control. A diet deficiency in methionine affects digestibility of essential amino acids and cysteine, but not the digestibility of methionine. The change in digestibility is reflected in the mRNA expression of amino acid transporters across different tissues.
Collapse
Affiliation(s)
- Naiara S Fagundes
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602; Department of Animal Science, University of Sao Paulo, Piracicaba, Sao Paulo, Brazil
| | - Marie C Milfort
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - Susan M Williams
- Department of Population Health, University of Georgia, Athens, GA 30602
| | - Manuel J Da Costa
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - Alberta L Fuller
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - José F Menten
- Department of Animal Science, University of Sao Paulo, Piracicaba, Sao Paulo, Brazil
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602
| | - Samuel E Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602.
| |
Collapse
|
20
|
To VPTH, Masagounder K, Loewen ME. SLC transporters ASCT2, B 0 AT1-like, y + LAT1, and LAT4-like associate with methionine electrogenic and radio-isotope flux kinetics in rainbow trout intestine. Physiol Rep 2019; 7:e14274. [PMID: 31705630 PMCID: PMC6841986 DOI: 10.14814/phy2.14274] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/21/2019] [Indexed: 01/08/2023] Open
Abstract
Methionine (Met) is an important building block and metabolite for protein biosynthesis. However, the mechanism behind its absorption in the fish gut has not been elucidated. Here, we describe the fundamental properties of Met transport along trout gut at µmol/L and mmol/L concentration. Both electrogenic and unidirectional DL-[14 C]Met flux were employed to characterize Met transporters in Ussing chambers. Exploiting the differences in gene expression between diploid (2N) and triploid (3N) and intestinal segment as tools, allowed the association between gene and methionine transport. Specifically, three intestinal segments including pyloric caeca (PC), midgut (MG), and hindgut (HG) were assessed. Results at 0-150 µmol/L concentration demonstrated that the DL-Met was most likely transported by apical transporter ASCT2 (SLC1A5) and recycled by basolateral transporter y+ LAT1 (SLC7A7) due to five lines of observation: (1) lack of Na+ -independent kinetics, (2) low expression of B0 AT2-like gene, (3) Na+ -dependent, high-affinity (Km , µmol/L ranges) kinetics in DL-[14 C]Met flux, (4) association mRNA expression with the high-affinity kinetics and (5) electrogenic currents induced by Met. Results at 0.2-20 mmol/L concentration suggested that the DL-Met transport is likely transported by B0 AT1-like (SLC6A19-like) based on gene expression, Na+ -dependence and low-affinity kinetics (Km , mmol/L ranges). Similarly, genomic and gene expression analysis suggest that the basolateral exit of methionine was primarily through LAT4-like transporter (SLC43A2-like). Conclusively, DL-Met uptake in trout gut was most likely governed by Na+ -dependent apical transporters ASCT2 and B0 AT1-like and released through basolateral LAT4-like, with some recycling through y+ LAT1. A comparatively simpler model than that previously described in mammals.
Collapse
Affiliation(s)
- Van P. T. H. To
- Veterinary Biomedical SciencesUniversity of SaskatchewanSaskatoonSaskatchewanCanada
| | | | - Matthew E. Loewen
- Veterinary Biomedical SciencesUniversity of SaskatchewanSaskatoonSaskatchewanCanada
| |
Collapse
|
21
|
Abstract
Background Cystinuria is an inherited disorder of renal amino acid transport that causes recurrent nephrolithiasis and significant morbidity in humans. It has an incidence of 1 in 7000 worldwide making it one of the most common genetic disorders in man. We phenotypically characterized a mouse model of cystinuria type A resultant from knockout of Slc3a1. Methods Knockout of Slc3a1 at RNA and protein levels was evaluated using real-time quantitative PCR and immunofluorescence. Slc3a1 knockout mice were placed on normal or breeder chow diets and evaluated for cystine stone formation over time suing x-ray analysis, and the development of kidney injury by measuring injury biomarkers. Kidney injury was also evaluated via histologic analysis. Amino acid levels were measured in the blood of mice using high performance liquid chromatography. Liver glutathione levels were measured using a luminescent-based assay. Results We confirmed knockout of Slc3a1 at the RNA level, while Slc7a9 RNA representing the co-transporter was preserved. As expected, we observed bladder stone formation in Slc3a1−/− mice. Male Slc3a1−/− mice exhibited lower weights compared to Slc3a1+/+. Slc3a1−/− mice on a regular diet demonstrated elevated blood urea nitrogen (BUN) without elevation of serum creatinine. However, placing the knockout animals on a breeder chow diet, containing a higher cystine concentration, resulted in the development of elevation of both BUN and creatinine indicative of more severe chronic kidney disease. Histological examination revealed that these dietary effects resulted in worsened kidney tubular obstruction and interstitial inflammation as well as worsened bladder inflammation. Cystine is a precursor for the antioxidant molecule glutathione, so we evaluated glutathione levels in the livers of Slc3a1−/− mice. We found significantly lowered levels of both reduced and total glutathione in the knockout animals. Conclusions Our results suggest that that diet can affect the development and progression of chronic kidney disease in an animal model of cystinuria, which may have important implications for patients with this disease. Additionally, reduced glutathione may predispose those with cystinuria to injury caused by oxidative stress. Word count: 327. Electronic supplementary material The online version of this article (10.1186/s12882-019-1417-8) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Cystinuria: genetic aspects, mouse models, and a new approach to therapy. Urolithiasis 2018; 47:57-66. [PMID: 30515543 DOI: 10.1007/s00240-018-1101-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/27/2018] [Indexed: 01/07/2023]
Abstract
Cystinuria, a genetic disorder of cystine transport, is characterized by excessive excretion of cystine in the urine and recurrent cystine stones in the kidneys and, to a lesser extent, in the bladder. Males generally are more severely affected than females. The disorder may lead to chronic kidney disease in many patients. The cystine transporter (b0,+) is a heterodimer consisting of the rBAT (encoded by SLC3A1) and b0,+AT (encoded by SLC7A9) subunits joined by a disulfide bridge. The molecular basis of cystinuria is known in great detail, and this information is now being used to define genotype-phenotype correlations. Current treatments for cystinuria include increased fluid intake to increase cystine solubility and the administration of thiol drugs for more severe cases. These drugs, however, have poor patient compliance due to adverse effects. Thus, there is a need to reduce or eliminate the risks associated with therapy for cystinuria. Four mouse models for cystinuria have been described and these models provide a resource for evaluating the safety and efficacy of new therapies for cystinuria. We are evaluating a new approach for the treatment of cystine stones based on the inhibition of cystine crystal growth by cystine analogs. Our ongoing studies indicate that cystine diamides are effective in preventing cystine stone formation in the Slc3a1 knockout mouse model for cystinuria. In addition to crystal growth, crystal aggregation is required for stone formation. Male and female mice with cystinuria have comparable levels of crystalluria, but very few female mice form stones. The identification of factors that inhibit cystine crystal aggregation in female mice may provide insight into the gender difference in disease severity in patients with cystinuria.
Collapse
|
23
|
Sumayao R, Newsholme P, McMorrow T. The Role of Cystinosin in the Intermediary Thiol Metabolism and Redox Homeostasis in Kidney Proximal Tubular Cells. Antioxidants (Basel) 2018; 7:antiox7120179. [PMID: 30513914 PMCID: PMC6315507 DOI: 10.3390/antiox7120179] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/22/2018] [Accepted: 11/24/2018] [Indexed: 01/26/2023] Open
Abstract
Cystinosin is a lysosomal transmembrane protein which facilitates transport of the disulphide amino acid cystine (CySS) from the lysosomes of the cell. This protein is encoded by the CTNS gene which is defective in the lysosomal storage disorder, cystinosis. Because of the apparent involvement of cystinosin in the intermediary thiol metabolism, its discovery has fuelled investigations into its role in modulating cellular redox homeostasis. The kidney proximal tubular cells (PTCs) have become the focus of various studies on cystinosin since the protein is highly expressed in these cells and kidney proximal tubular transport dysfunction is the foremost clinical manifestation of cystinosis. The lysosomal CySS pool is a major source of cytosolic cysteine (Cys), the limiting amino acid for the synthesis of an important antioxidant glutathione (GSH) via the γ-glutamyl cycle. Therefore, loss of cystinosin function is presumed to lead to cytosolic deficit of Cys which may impair GSH synthesis. However, studies using in vitro models lacking cystinosin yielded inconsistent results and failed to establish the mechanistic role of cystinosin in modulating GSH synthesis and redox homeostasis. Because of the complexity of the metabolic micro- and macro-environment in vivo, using in vitro models alone may not be able to capture the complete sequence of biochemical and physiological events that occur as a consequence of loss of cystinosin function. The coexistence of pathways for the overall handling and disposition of GSH, the modulation of CTNS gene by intracellular redox status and the existence of a non-canonical isoform of cystinosin may constitute possible rescue mechanisms in vivo to remediate redox perturbations in renal PTCs. Importantly, the mitochondria seem to play a critical role in orchestrating redox imbalances initiated by cystinosin dysfunction. Non-invasive techniques such as in vivo magnetic resonance imaging with the aid of systems biology approaches may provide invaluable mechanistic insights into the role of cystinosin in the essential intermediary thiol metabolism and in the overall regulation cellular redox homeostasis.
Collapse
Affiliation(s)
- Rodolfo Sumayao
- Chemistry Department, De La Salle University, Manila 1004, Philippines.
| | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences and Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth 6845, Australia.
| | - Tara McMorrow
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
24
|
Akanuma SI, Yamakoshi A, Sugouchi T, Kubo Y, Hartz AMS, Bauer B, Hosoya KI. Role of l-Type Amino Acid Transporter 1 at the Inner Blood-Retinal Barrier in the Blood-to-Retina Transport of Gabapentin. Mol Pharm 2018; 15:2327-2337. [PMID: 29688723 DOI: 10.1021/acs.molpharmaceut.8b00179] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gabapentin is an antiseizure drug that is known to also have beneficial effects on the retinal cells. To use gabapentin in retinal pharmacotherapy, it is critical to understand gabapentin distribution in the retina. The purpose of this study was to clarify the kinetics of gabapentin influx transport across the inner and outer blood-retinal barrier (BRB), which regulates the exchange of compounds/drugs between the circulating blood and the retina. In vivo blood-to-retina gabapentin transfer was evaluated by the rat carotid artery injection technique. In addition, gabapentin transport was examined using in vitro models of the inner (TR-iBRB2 cells) and outer BRB (RPE-J cells). The in vivo [3H]gabapentin transfer to the rat retina across the BRB was significantly reduced in the presence of unlabeled gabapentin, suggesting transporter-mediated blood-to-retina distribution of gabapentin. Substrates of the Na+-independent l-type amino acid transporter 1 (LAT1), such as 2-aminobicyclo[2.2.1]heptane-2-carboxylic acid (BCH), also significantly inhibited the in vivo [3H]gabapentin transfer. [3H]Gabapentin uptake in TR-iBRB2 and RPE-J cells exhibited Na+-independent and saturable kinetics with a Km of 735 and 507 μM, respectively. Regarding the effect of various transporter substrates/inhibitors on gabapentin transport in these cells, LAT1 substrates significantly inhibited [3H]gabapentin uptake in TR-iBRB2 and RPE-J cells. In addition, preloaded [3H]gabapentin release from TR-iBRB2 and RPE-J cells was trans-stimulated by LAT1 substrates through the obligatory exchange mechanism as LAT1. Immunoblot analysis indicates the protein expression of LAT1 in TR-iBRB2 and RPE-J cells. These results imply that LAT1 at the inner and outer BRB takes part in gabapentin transport between the circulating blood and retina. Moreover, treatment of LAT1-targeted small interfering RNA to TR-iBRB2 cells significantly reduced both the level of LAT1 protein expression and [3H]gabapentin uptake activities in TR-iBRB2 cells. In conclusion, data from the present study indicate that LAT1 at the inner BRB is involved in retinal gabapentin transfer, and also suggest that LAT1 mediates gabapentin transport in the RPE cells.
Collapse
Affiliation(s)
- Shin-Ichi Akanuma
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| | - Atsuko Yamakoshi
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| | - Takeshi Sugouchi
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| | - Yoshiyuki Kubo
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| | - Anika M S Hartz
- Sanders-Brown Center on Aging , University of Kentucky , Lexington , KY 40536 , United States.,Department of Pharmacology and Nutritional Sciences, College of Medicine , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Ken-Ichi Hosoya
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , Sugitani , 2630 Toyama , Japan
| |
Collapse
|
25
|
Affiliation(s)
- Scott V. Wiener
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
| | - Thomas Chi
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
| | - Marshall L Stoller
- Department of Urology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
26
|
Martell HJ, Wong KA, Martin JF, Kassam Z, Thomas K, Wass MN. Associating mutations causing cystinuria with disease severity with the aim of providing precision medicine. BMC Genomics 2017; 18:550. [PMID: 28812535 PMCID: PMC5558187 DOI: 10.1186/s12864-017-3913-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Cystinuria is an inherited disease that results in the formation of cystine stones in the kidney, which can have serious health complications. Two genes (SLC7A9 and SLC3A1) that form an amino acid transporter are known to be responsible for the disease. Variants that cause the disease disrupt amino acid transport across the cell membrane, leading to the build-up of relatively insoluble cystine, resulting in formation of stones. Assessing the effects of each mutation is critical in order to provide tailored treatment options for patients. We used various computational methods to assess the effects of cystinuria associated mutations, utilising information on protein function, evolutionary conservation and natural population variation of the two genes. We also analysed the ability of some methods to predict the phenotypes of individuals with cystinuria, based on their genotypes, and compared this to clinical data. Results Using a literature search, we collated a set of 94 SLC3A1 and 58 SLC7A9 point mutations known to be associated with cystinuria. There are differences in sequence location, evolutionary conservation, allele frequency, and predicted effect on protein function between these mutations and other genetic variants of the same genes that occur in a large population. Structural analysis considered how these mutations might lead to cystinuria. For SLC7A9, many mutations swap hydrophobic amino acids for charged amino acids or vice versa, while others affect known functional sites. For SLC3A1, functional information is currently insufficient to make confident predictions but mutations often result in the loss of hydrogen bonds and largely appear to affect protein stability. Finally, we showed that computational predictions of mutation severity were significantly correlated with the disease phenotypes of patients from a clinical study, despite different methods disagreeing for some of their predictions. Conclusions The results of this study are promising and highlight the areas of research which must now be pursued to better understand how mutations in SLC3A1 and SLC7A9 cause cystinuria. The application of our approach to a larger data set is essential, but we have shown that computational methods could play an important role in designing more effective personalised treatment options for patients with cystinuria. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3913-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Henry J Martell
- School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| | - Kathie A Wong
- Urology Centre, Guy's and St. Thomas' NHS Foundation Trust, London, SE1 9RT, UK
| | - Juan F Martin
- School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| | - Ziyan Kassam
- Urology Centre, Guy's and St. Thomas' NHS Foundation Trust, London, SE1 9RT, UK
| | - Kay Thomas
- Urology Centre, Guy's and St. Thomas' NHS Foundation Trust, London, SE1 9RT, UK.
| | - Mark N Wass
- School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NJ, UK.
| |
Collapse
|
27
|
Mizukami K, Raj K, Osborne C, Giger U. Cystinuria Associated with Different SLC7A9 Gene Variants in the Cat. PLoS One 2016; 11:e0159247. [PMID: 27404572 PMCID: PMC4942060 DOI: 10.1371/journal.pone.0159247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/29/2016] [Indexed: 02/03/2023] Open
Abstract
Cystinuria is a classical inborn error of metabolism characterized by a selective proximal renal tubular defect affecting cystine, ornithine, lysine, and arginine (COLA) reabsorption, which can lead to uroliths and urinary obstruction. In humans, dogs and mice, cystinuria is caused by variants in one of two genes, SLC3A1 and SLC7A9, which encode the rBAT and bo,+AT subunits of the bo,+ basic amino acid transporter system, respectively. In this study, exons and flanking regions of the SLC3A1 and SLC7A9 genes were sequenced from genomic DNA of cats (Felis catus) with COLAuria and cystine calculi. Relative to the Felis catus-6.2 reference genome sequence, DNA sequences from these affected cats revealed 3 unique homozygous SLC7A9 missense variants: one in exon 5 (p.Asp236Asn) from a non-purpose-bred medium-haired cat, one in exon 7 (p.Val294Glu) in a Maine Coon and a Sphinx cat, and one in exon 10 (p.Thr392Met) from a non-purpose-bred long-haired cat. A genotyping assay subsequently identified another cystinuric domestic medium-haired cat that was homozygous for the variant originally identified in the purebred cats. These missense variants result in deleterious amino acid substitutions of highly conserved residues in the bo,+AT protein. A limited population survey supported that the variants found were likely causative. The remaining 2 sequenced domestic short-haired cats had a heterozygous variant at a splice donor site in intron 10 and a homozygous single nucleotide variant at a branchpoint in intron 11 of SLC7A9, respectively. This study identifies the first SLC7A9 variants causing feline cystinuria and reveals that, as in humans and dogs, this disease is genetically heterogeneous in cats.
Collapse
Affiliation(s)
- Keijiro Mizukami
- Section of Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Karthik Raj
- Section of Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Carl Osborne
- Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States of America
| | - Urs Giger
- Section of Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
28
|
Novel cystine transporter in renal proximal tubule identified as a missing partner of cystinuria-related plasma membrane protein rBAT/SLC3A1. Proc Natl Acad Sci U S A 2016; 113:775-80. [PMID: 26739563 DOI: 10.1073/pnas.1519959113] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Heterodimeric amino acid transporters play crucial roles in epithelial transport, as well as in cellular nutrition. Among them, the heterodimer of a membrane protein b(0,+)AT/SLC7A9 and its auxiliary subunit rBAT/SLC3A1 is responsible for cystine reabsorption in renal proximal tubules. The mutations in either subunit cause cystinuria, an inherited amino aciduria with impaired renal reabsorption of cystine and dibasic amino acids. However, an unsolved paradox is that rBAT is highly expressed in the S3 segment, the late proximal tubules, whereas b(0,+)AT expression is highest in the S1 segment, the early proximal tubules, so that the presence of an unknown partner of rBAT in the S3 segment has been proposed. In this study, by means of coimmunoprecipitation followed by mass spectrometry, we have found that a membrane protein AGT1/SLC7A13 is the second partner of rBAT. AGT1 is localized in the apical membrane of the S3 segment, where it forms a heterodimer with rBAT. Depletion of rBAT in mice eliminates the expression of AGT1 in the renal apical membrane. We have reconstituted the purified AGT1-rBAT heterodimer into proteoliposomes and showed that AGT1 transports cystine, aspartate, and glutamate. In the apical membrane of the S3 segment, AGT1 is suggested to locate itself in close proximity to sodium-dependent acidic amino acid transporter EAAC1 for efficient functional coupling. EAAC1 is proposed to take up aspartate and glutamate released into luminal fluid by AGT1 due to its countertransport so that preventing the urinary loss of aspartate and glutamate. Taken all together, AGT1 is the long-postulated second cystine transporter in the S3 segment of proximal tubules and a possible candidate to be involved in isolated cystinuria.
Collapse
|
29
|
Mercury toxicokinetics of the healthy human term placenta involve amino acid transporters and ABC transporters. Toxicology 2016; 340:34-42. [DOI: 10.1016/j.tox.2015.12.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/10/2015] [Accepted: 12/23/2015] [Indexed: 11/18/2022]
|
30
|
The role of N-glycans and the C-terminal loop of the subunit rBAT in the biogenesis of the cystinuria-associated transporter. Biochem J 2015; 473:233-44. [PMID: 26537754 DOI: 10.1042/bj20150846] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/03/2015] [Indexed: 11/17/2022]
Abstract
The transport system b(0,+) mediates reabsorption of dibasic amino acids and cystine in the kidney. It is made up of two disulfide-linked membrane subunits: the carrier, b(0,+)AT and the helper, rBAT (related to b(0,+) amino acid transporter). rBAT mutations that impair biogenesis of the transporter cause type I cystinuria. It has been shown that upon assembly, b(0,+)AT prevents degradation and promotes folding of rBAT; then, rBAT traffics b(0,+)AT from the endoplasmic reticulum (ER) to the plasma membrane. The role of the N-glycans of rBAT and of its C-terminal loop, which has no homology to any other sequence, in biogenesis of system b(0,+) is unknown. In the present study, we studied these points. We first identified the five N-glycans of rBAT. Elimination of the N-glycan Asn(575), but not of the others, delayed transporter maturation, as measured by pulse chase experiments and endoglycosidase H assays. Moreover, a transporter with only the N-glycan Asn(575) displayed similar maturation compared with wild-type, suggesting that this N-glycan was necessary and sufficient to achieve the maximum rate of transporter maturation. Deletion of the rBAT C-terminal disulfide loop (residues 673-685) prevented maturation and prompted degradation of the transporter. Alanine-scanning mutagenesis uncovered loop residues important for stability and/or maturation of system b(0,+). Further, double-mutant cycle analysis showed partial additivity of the effects of the Asn(679) loop residue and the N-glycan Asn(575) on transporter maturation, indicating that they may interact during system b(0,+) biogenesis. These data highlight the important role of the N-glycan Asn(575) and the C-terminal disulfide loop of rBAT in biogenesis of the rBAT-b(0,+)AT heterodimer.
Collapse
|
31
|
Abstract
Cystinuria is an aminoaciduria caused by mutations in the genes that encode the two subunits of the amino acid transport system b0,+, responsible for the renal reabsorption of cystine and dibasic amino acids. The clinical symptoms of cystinuria relate to nephrolithiasis, due to the precipitation of cystine in urine. Mutations in SLC3A1, which codes for the heavy subunit rBAT, cause cystinuria type A, whereas mutations in SLC7A9, which encodes the light subunit b0,+AT, cause cystinuria type B. By crossing Slc3a1-/- with Slc7a9-/- mice we generated a type AB cystinuria mouse model to test digenic inheritance of cystinuria. The 9 genotypes obtained have been analyzed at early (2- and 5-months) and late stage (8-months) of the disease. Monitoring the lithiasic phenotype by X-ray, urine amino acid content analysis and protein expression studies have shown that double heterozygous mice (Slc7a9+/-Slc3a1+/-) present lower expression of system b0,+ and higher hyperexcretion of cystine than single heterozygotes (Slc7a9+/-Slc3a1+/+ and Slc7a9+/+Slc3a1+/-) and give rise to lithiasis in 4% of the mice, demonstrating that cystinuria has a digenic inheritance in this mouse model. Moreover in this study it has been demonstrated a genotype/phenotype correlation in type AB cystinuria mouse model providing new insights for further molecular and genetic studies of cystinuria patients.
Collapse
|
32
|
Vuille-dit-Bille RN, Camargo SM, Emmenegger L, Sasse T, Kummer E, Jando J, Hamie QM, Meier CF, Hunziker S, Forras-Kaufmann Z, Kuyumcu S, Fox M, Schwizer W, Fried M, Lindenmeyer M, Götze O, Verrey F. Human intestine luminal ACE2 and amino acid transporter expression increased by ACE-inhibitors. Amino Acids 2014; 47:693-705. [DOI: 10.1007/s00726-014-1889-6] [Citation(s) in RCA: 229] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/03/2014] [Indexed: 02/06/2023]
|
33
|
Mizukami K, Raj K, Giger U. Feline cystinuria caused by a missense mutation in the SLC3A1 gene. J Vet Intern Med 2014; 29:120-5. [PMID: 25417848 PMCID: PMC4858075 DOI: 10.1111/jvim.12501] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/17/2014] [Accepted: 10/13/2014] [Indexed: 11/29/2022] Open
Abstract
Background Cystinuria is an inherited metabolic disease that is relatively common in dogs, but rare in cats and is characterized by defective amino acid reabsorption, leading to cystine urolithiasis. Objectives The aim of this study was to report on a mutation in a cystinuric cat. Animals A male domestic shorthair (DSH) cat with cystine calculi, 11 control cats from Wyoming, and 54 DSH and purebred control cats from elsewhere in the United States. Methods Exons of the SLC3A1 gene were sequenced from genomic DNA of the cystinuric cat and a healthy cat. Genetic screening for the discovered polymorphisms was conducted on all cats. Results A DSH cat showed stranguria beginning at 2 months of age, and cystine calculi were removed at 4 months of age. The cat was euthanized at 6 months of age because of neurological signs possibly related to arginine deficiency. Twenty‐five SLC3A1 polymorphisms were observed in the sequenced cats when compared to the feline reference sequence. The cystinuric cat was homozygous for 5 exonic and 8 noncoding SLC3A1 polymorphisms, and 1 of them was a unique missense mutation (c.1342C>T). This mutation results in a deleterious amino acid substitution (p.Arg448Trp) of a highly conserved arginine residue in the rBAT protein encoded by the SLC3A1 gene. This mutation was found previously in cystinuric human patients, but was not seen in any other tested cats. Conclusions and Clinical Importance This study is the first report of an SLC3A1 mutation causing cystinuria in a cat, and could be used to characterize other cystinuric cats at the molecular level.
Collapse
Affiliation(s)
- K Mizukami
- Section of Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | | | | |
Collapse
|
34
|
Edvardsson VO, Goldfarb DS, Lieske JC, Beara-Lasic L, Anglani F, Milliner DS, Palsson R. Hereditary causes of kidney stones and chronic kidney disease. Pediatr Nephrol 2013; 28:1923-42. [PMID: 23334384 PMCID: PMC4138059 DOI: 10.1007/s00467-012-2329-z] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 09/13/2012] [Accepted: 09/17/2012] [Indexed: 01/27/2023]
Abstract
Adenine phosphoribosyltransferase (APRT) deficiency, cystinuria, Dent disease, familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHHNC), and primary hyperoxaluria (PH) are rare but important causes of severe kidney stone disease and/or chronic kidney disease in children. Recurrent kidney stone disease and nephrocalcinosis, particularly in pre-pubertal children, should alert the physician to the possibility of an inborn error of metabolism as the underlying cause. Unfortunately, the lack of recognition and knowledge of the five disorders has frequently resulted in an unacceptable delay in diagnosis and treatment, sometimes with grave consequences. A high index of suspicion coupled with early diagnosis may reduce or even prevent the serious long-term complications of these diseases. In this paper, we review the epidemiology, clinical features, diagnosis, treatment, and outcome of patients with APRT deficiency, cystinuria, Dent disease, FHHNC, and PH, with an emphasis on childhood manifestations.
Collapse
MESH Headings
- Adenine Phosphoribosyltransferase/deficiency
- Adenine Phosphoribosyltransferase/genetics
- Animals
- Child
- Cystinuria/diagnosis
- Cystinuria/epidemiology
- Cystinuria/genetics
- Cystinuria/therapy
- Dent Disease/diagnosis
- Dent Disease/epidemiology
- Dent Disease/genetics
- Dent Disease/therapy
- Genetic Predisposition to Disease
- Heredity
- Humans
- Hypercalciuria/diagnosis
- Hypercalciuria/epidemiology
- Hypercalciuria/genetics
- Hypercalciuria/therapy
- Hyperoxaluria, Primary/diagnosis
- Hyperoxaluria, Primary/epidemiology
- Hyperoxaluria, Primary/genetics
- Hyperoxaluria, Primary/therapy
- Kidney Calculi/diagnosis
- Kidney Calculi/epidemiology
- Kidney Calculi/genetics
- Kidney Calculi/therapy
- Metabolism, Inborn Errors/diagnosis
- Metabolism, Inborn Errors/epidemiology
- Metabolism, Inborn Errors/genetics
- Metabolism, Inborn Errors/therapy
- Nephrocalcinosis/diagnosis
- Nephrocalcinosis/epidemiology
- Nephrocalcinosis/genetics
- Nephrocalcinosis/therapy
- Phenotype
- Prognosis
- Renal Insufficiency, Chronic/diagnosis
- Renal Insufficiency, Chronic/epidemiology
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/therapy
- Renal Tubular Transport, Inborn Errors/diagnosis
- Renal Tubular Transport, Inborn Errors/epidemiology
- Renal Tubular Transport, Inborn Errors/genetics
- Renal Tubular Transport, Inborn Errors/therapy
- Risk Factors
- Urolithiasis/diagnosis
- Urolithiasis/epidemiology
- Urolithiasis/genetics
- Urolithiasis/therapy
Collapse
|
35
|
Brons AK, Henthorn PS, Raj K, Fitzgerald CA, Liu J, Sewell AC, Giger U. SLC3A1 and SLC7A9 mutations in autosomal recessive or dominant canine cystinuria: a new classification system. J Vet Intern Med 2013; 27:1400-8. [PMID: 24001348 DOI: 10.1111/jvim.12176] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 07/11/2013] [Accepted: 07/24/2013] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Cystinuria, one of the first recognized inborn errors of metabolism, has been reported in many dog breeds. HYPOTHESIS/OBJECTIVES To determine urinary cystine concentrations, inheritance, and mutations in the SLC3A1 and SLC7A9 genes associated with cystinuria in 3 breeds. ANIMALS Mixed and purebred Labrador Retrievers (n = 6), Australian Cattle Dogs (6), Miniature Pinschers (4), and 1 mixed breed dog with cystine urolithiasis, relatives and control dogs. METHODS Urinary cystinuria and aminoaciduria was assessed and exons of the SLC3A1 and SLC7A9 genes were sequenced from genomic DNA. RESULTS In each breed, male and female dogs, independent of neuter status, were found to form calculi. A frameshift mutation in SLC3A1 (c.350delG) resulting in a premature stop codon was identified in autosomal-recessive (AR) cystinuria in Labrador Retrievers and mixed breed dogs. A 6 bp deletion (c.1095_1100del) removing 2 threonines in SLC3A1 was found in autosomal-dominant (AD) cystinuria with a more severe phenotype in homozygous than in heterozygous Australian Cattle Dogs. A missense mutation in SLC7A9 (c.964G>A) was discovered in AD cystinuria in Miniature Pinschers with only heterozygous affected dogs observed to date. Breed-specific DNA tests were developed, but the prevalence of each mutation remains unknown. CONCLUSIONS AND CLINICAL IMPORTANCE These studies describe the first AD inheritance and the first putative SLC7A9 mutation to cause cystinuria in dogs and expand our understanding of this phenotypically and genetically heterogeneous disease, leading to a new classification system for canine cystinuria and better therapeutic management and genetic control in these breeds.
Collapse
Affiliation(s)
- A-K Brons
- Section of Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | | | | |
Collapse
|
36
|
Schweikhard ES, Ziegler CM. Amino acid secondary transporters: toward a common transport mechanism. CURRENT TOPICS IN MEMBRANES 2013. [PMID: 23177982 DOI: 10.1016/b978-0-12-394316-3.00001-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Solute carriers (SLC) that transport amino acids are key players in health and diseases in humans. Their prokaryotic relatives are often involved in essential physiological processes in microorganisms, e.g. in homeostasis and acidic/osmotic stress response. High-resolution X-ray structures of the sequence-unrelated amino acid transporters unraveled a striking structural similarity between carriers, which were formerly assigned to different families. The highly conserved fold is characterized by two inverted structural repeats of five transmembrane helices each and indicates common mechanistic transport concepts if not an evolutionary link among a large number of amino acid transporters. Therefore, these transporters are classified now into the structural amino acid-polyamine-organocation superfamily (APCS). The APCS includes among others the mammalian SLC6 transporters and the heterodimeric SLC7/SLC3 transporters. However, it has to be noted that the APCS is not limited entirely to amino acid transporters but contains also transporters for, e.g. amino acid derivatives and sugars. For instance, the betaine-choline-carnitine transporter family of bacterial activity-regulated Na(+)- and H(+)-coupled symporters for glycine betaine and choline is also part of this second largest structural superfamily. The APCS fold provides different possibilities to transport the same amino acid. Arginine can be transported by an H(+)-coupled symport or by antiport mechanism in exchange against agmatine for example. The convergence of the mechanistic concept of transport under comparable physiological conditions allows speculating if structurally unexplored amino acid transporters, e.g. the members of the SLC36 and SLC38 family, belong to the APCS, too. In the kidney, which is an organ that depends critically on the regulated amino acid transport, these different SLC transporters have to work together to account for proper function. Here, we will summarize the basic concepts of Na(+)- and H(+)-coupled amino acid symport and amino acid-product antiport in the light of the respective physiological requirements.
Collapse
Affiliation(s)
- Eva S Schweikhard
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt, Germany
| | | |
Collapse
|
37
|
Expression of human heteromeric amino acid transporters in the yeast Pichia pastoris. Protein Expr Purif 2012; 87:35-40. [PMID: 23085088 DOI: 10.1016/j.pep.2012.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 10/08/2012] [Accepted: 10/08/2012] [Indexed: 11/20/2022]
Abstract
Human heteromeric amino acid transporters (HATs) play key roles in renal and intestinal re-absorption, cell redox balance and tumor growth. These transporters are composed of a heavy and a light subunit, which are connected by a disulphide bridge. Heavy subunits are the two type II membrane N-glycoproteins rBAT and 4F2hc, while L-type amino acid transporters (LATs) are the light and catalytic subunits of HATs. We tested the expression of human 4F2hc and rBAT as well as seven light subunits in the methylotrophic yeast Pichia pastoris. 4F2hc and the light subunit LAT2 showed the highest expression levels and yields after detergent solubilization. Co-transformation of both subunits in Pichia cells resulted in overexpression of the disulphide bridge-linked 4F2hc/LAT2 heterodimer. Two sequential affinity chromatography steps were applied to purify detergent-solubilized heterodimers yielding ~1mg of HAT from 2l of cell culture. Our results indicate that P. pastoris is a convenient system for the expression and purification of human 4F2hc/LAT2 for structural studies.
Collapse
|
38
|
Rius M, Chillarón J. Carrier subunit of plasma membrane transporter is required for oxidative folding of its helper subunit. J Biol Chem 2012; 287:18190-200. [PMID: 22493502 DOI: 10.1074/jbc.m111.321943] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We study the amino acid transport system b(0,+) as a model for folding, assembly, and early traffic of membrane protein complexes. System b(0,+) is made of two disulfide-linked membrane subunits: the carrier, b(0,+) amino acid transporter (b(0,+)AT), a polytopic protein, and the helper, related to b(0,+) amino acid transporter (rBAT), a type II glycoprotein. rBAT ectodomain mutants display folding/trafficking defects that lead to type I cystinuria. Here we show that, in the presence of b(0,+)AT, three disulfides were formed in the rBAT ectodomain. Disulfides Cys-242-Cys-273 and Cys-571-Cys-666 were essential for biogenesis. Cys-673-Cys-685 was dispensable, but the single mutants C673S, and C685S showed compromised stability and trafficking. Cys-242-Cys-273 likely was the first disulfide to form, and unpaired Cys-242 or Cys-273 disrupted oxidative folding. Strikingly, unassembled rBAT was found as an ensemble of different redox species, mainly monomeric. The ensemble did not change upon inhibition of rBAT degradation. Overall, these results indicated a b(0,+)AT-dependent oxidative folding of the rBAT ectodomain, with the initial and probably cotranslational formation of Cys-242-Cys-273, followed by the oxidation of Cys-571-Cys-666 and Cys-673-Cys-685, that was completed posttranslationally.
Collapse
Affiliation(s)
- Mònica Rius
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, E-08028 Barcelona, Spain
| | | |
Collapse
|
39
|
Heat-shock mediated overexpression of HNF1β mutations has differential effects on gene expression in the Xenopus pronephric kidney. PLoS One 2012; 7:e33522. [PMID: 22438943 PMCID: PMC3305329 DOI: 10.1371/journal.pone.0033522] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 02/15/2012] [Indexed: 02/07/2023] Open
Abstract
The transcription factor HNF1B, encoded by the TCF2 gene, plays an important role in the organogenesis of vertebrates. In humans, heterozygous mutations of HNF1B are associated with several diseases, such as pancreatic β-cell dysfunction leading to maturity-onset diabetes of the young (MODY5), defective kidney development, disturbed liver function, pancreas atrophy, and malformations of the genital tract. The African claw frog Xenopus laevis is an excellent model to study the processes involved in embryogenesis and organogenesis, as it can be manipulated easily with a series of methods. In the present study, we overexpressed HNF1β mutants in the developing Xenopus embryo to assess their roles during organogenesis, particularly in the developing pronephric kidney. Towards this goal, we developed a heat-shock inducible binary Cre/loxP system with activator and effector strains. Heat-shock activation of the mutant HNF1B variants P328L329del and A263insGG resulted in malformations of various organs and the affected larvae developed large edemas. Defects in the pronephros were primarily confined to malformed proximal tubules. Furthermore, the expression of the proximal tubule marker genes tmem27 and slc3a1, both involved in amino acid transport, was affected. Both P328L329del and A263insGG downregulated expression of slc3a1. In addition, P328L329del reduced tmem27 expression while A263insGG overexpression decreased expression of the chloride channel clcnk and the transcription factor pax2. Overexpression of two mutant HNF1B derivatives resulted in distinct phenotypes reflected by either a reduction or an enlargement of pronephros size. The expression of selected pronephric marker genes was differentially affected upon overexpression of HNF1B mutations. Based on our findings, we postulate that HNF1B mutations influence gene regulation upon overexpression in specific and distinct manners. Furthermore, our study demonstrates that the newly established Cre/loxP system for Xenopus embryos is an attractive alternative to examine the gene regulatory potential of transcription factors in developing pronephric kidney as exemplified here for HNF1B.
Collapse
|
40
|
Sebastio G, Sperandeo MP, Andria G. Lysinuric protein intolerance: reviewing concepts on a multisystem disease. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2011; 157C:54-62. [PMID: 21308987 DOI: 10.1002/ajmg.c.30287] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lysinuric protein intolerance (LPI) is an inherited aminoaciduria caused by defective cationic amino acid transport at the basolateral membrane of epithelial cells in intestine and kidney. LPI is caused by mutations in the SLC7A7 gene, which encodes the y(+)LAT-1 protein, the catalytic light chain subunit of a complex belonging to the heterodimeric amino acid transporter family. LPI was initially described in Finland, but has worldwide distribution. Typically, symptoms begin after weaning with refusal of feeding, vomiting, and consequent failure to thrive. Hepatosplenomegaly, hematological anomalies, neurological involvement, including hyperammonemic coma are recurrent clinical features. Two major complications, pulmonary alveolar proteinosis and renal disease are increasingly observed in LPI patients. There is extreme variability in the clinical presentation even within individual families, frequently leading to misdiagnosis or delayed diagnosis. This condition is diagnosed by urine amino acids, showing markedly elevated excretion of lysine and other dibasic amino acids despite low plasma levels of lysine, ornithine, and arginine. The biochemical diagnosis can be uncertain, requiring confirmation by DNA testing. So far, approximately 50 different mutations have been identified in the SLC7A7 gene in a group of 142 patients from 110 independent families. No genotype-phenotype correlation could be established. Therapy requires a low protein diet, low-dose citrulline supplementation, nitrogen-scavenging compounds to prevent hyperammonemia, lysine, and carnitine supplements. Supportive therapy is available for most complications with bronchoalveolar lavage being necessary for alveolar proteinosis.
Collapse
|
41
|
|
42
|
Raciti D, Reggiani L, Geffers L, Jiang Q, Bacchion F, Subrizi AE, Clements D, Tindal C, Davidson DR, Kaissling B, Brändli AW. Organization of the pronephric kidney revealed by large-scale gene expression mapping. Genome Biol 2008; 9:R84. [PMID: 18492243 PMCID: PMC2441470 DOI: 10.1186/gb-2008-9-5-r84] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 03/19/2008] [Accepted: 05/20/2008] [Indexed: 11/28/2022] Open
Abstract
Gene expression mapping reveals 8 functionally distinct domains in the Xenopus pronephros. Interestingly, no structure equivalent to the mammalian collecting duct is identified. Background The pronephros, the simplest form of a vertebrate excretory organ, has recently become an important model of vertebrate kidney organogenesis. Here, we elucidated the nephron organization of the Xenopus pronephros and determined the similarities in segmentation with the metanephros, the adult kidney of mammals. Results We performed large-scale gene expression mapping of terminal differentiation markers to identify gene expression patterns that define distinct domains of the pronephric kidney. We analyzed the expression of over 240 genes, which included members of the solute carrier, claudin, and aquaporin gene families, as well as selected ion channels. The obtained expression patterns were deposited in the searchable European Renal Genome Project Xenopus Gene Expression Database. We found that 112 genes exhibited highly regionalized expression patterns that were adequate to define the segmental organization of the pronephric nephron. Eight functionally distinct domains were discovered that shared significant analogies in gene expression with the mammalian metanephric nephron. We therefore propose a new nomenclature, which is in line with the mammalian one. The Xenopus pronephric nephron is composed of four basic domains: proximal tubule, intermediate tubule, distal tubule, and connecting tubule. Each tubule may be further subdivided into distinct segments. Finally, we also provide compelling evidence that the expression of key genes underlying inherited renal diseases in humans has been evolutionarily conserved down to the level of the pronephric kidney. Conclusion The present study validates the Xenopus pronephros as a genuine model that may be used to elucidate the molecular basis of nephron segmentation and human renal disease.
Collapse
Affiliation(s)
- Daniela Raciti
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Wolfgang-Pauli-Strasse 10, CH-8093 Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bartoccioni P, Rius M, Zorzano A, Palacín M, Chillarón J. Distinct classes of trafficking rBAT mutants cause the type I cystinuria phenotype. Hum Mol Genet 2008; 17:1845-54. [PMID: 18332091 DOI: 10.1093/hmg/ddn080] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Most mutations in the rBAT subunit of the heterodimeric cystine transporter rBAT-b(0,+)AT cause type I cystinuria. Trafficking of the transporter requires the intracellular assembly of the two subunits. Without its partner, rBAT, but not b(0,+)AT, is rapidly degraded. We analyzed the initial biogenesis of wild-type rBAT and type I cystinuria rBAT mutants. rBAT was degraded, at least in part, via the ERAD pathway. Assembly with b(0,+)AT within the endoplasmic reticulum (ER) blocked rBAT degradation and could be independent of the calnexin chaperone system. This system was, however, necessary for post-assembly maturation of the heterodimer. Without b(0,+)AT, wild-type and rBAT mutants were degraded with similar kinetics. In its presence, rBAT mutants showed strongly reduced (L89P) or no transport activity, failed to acquire complex N-glycosylation and to oligomerize, suggesting assembly and/or folding defects. Most of the transmembrane domain mutant L89P did not heterodimerize with b(0,+)AT and was degraded. However, the few [L89P]rBAT-b(0,+)AT heterodimers were stable, consistent with assembly, but not folding, defects. Mutants of the rBAT extracellular domain (T216M, R365W, M467K and M467T) efficiently assembled with b(0,+)AT but were subsequently degraded. Together with earlier results, the data suggest a two-step biogenesis model, with the early assembly of the subunits followed by folding of the rBAT extracellular domain. Defects on either of these steps lead to the type I cystinuria phenotype.
Collapse
Affiliation(s)
- Paola Bartoccioni
- Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| | | | | | | | | |
Collapse
|
44
|
Bröer S. Amino acid transport across mammalian intestinal and renal epithelia. Physiol Rev 2008; 88:249-86. [PMID: 18195088 DOI: 10.1152/physrev.00018.2006] [Citation(s) in RCA: 632] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The transport of amino acids in kidney and intestine is critical for the supply of amino acids to all tissues and the homeostasis of plasma amino acid levels. This is illustrated by a number of inherited disorders affecting amino acid transport in epithelial cells, such as cystinuria, lysinuric protein intolerance, Hartnup disorder, iminoglycinuria, dicarboxylic aminoaciduria, and some other less well-described disturbances of amino acid transport. The identification of most epithelial amino acid transporters over the past 15 years allows the definition of these disorders at the molecular level and provides a clear picture of the functional cooperation between transporters in the apical and basolateral membranes of mammalian epithelial cells. Transport of amino acids across the apical membrane not only makes use of sodium-dependent symporters, but also uses the proton-motive force and the gradient of other amino acids to efficiently absorb amino acids from the lumen. In the basolateral membrane, antiporters cooperate with facilitators to release amino acids without depleting cells of valuable nutrients. With very few exceptions, individual amino acids are transported by more than one transporter, providing backup capacity for absorption in the case of mutational inactivation of a transport system.
Collapse
Affiliation(s)
- Stefan Bröer
- School of Biochemistry and Molecular Biology, Australian National University, Canberra, Australian Capital Territory, Australia.
| |
Collapse
|
45
|
Wendt-Nordahl G, Sagi S, Bolenz C, Alken P, Michel MS, Knoll T. Evaluation of cystine transport in cultured human kidney cells and establishment of cystinuria type I phenotype by antisense technology. ACTA ACUST UNITED AC 2007; 36:25-9. [PMID: 18074125 DOI: 10.1007/s00240-007-0127-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Accepted: 11/21/2007] [Indexed: 10/22/2022]
Abstract
Cystinuria is a rare hereditary disease resulting in recurrent stone formation and the need for repeated invasive interventions. So far, two responsible genes have been identified which encode the two transporters, rBAT and b(0,+)AT forming a heterodimer to transport cystine in proximal tubular cells (PTC) and whose defect results in increased excretion of cystine. A human cell line mimicing the phenotype of cystinuria in vitro is yet to be developed. Human kidney (HK)-2 is a PTC line derived from normal HK. After determining the presence of rBAT gene by RT-PCR and Western blot analysis, radioactively labeled cystine (S(35)) was used to evaluate the functional presence of the amino acid transport in HK-2 cells when cultured in vitro. To achieve a cystinuria type I phenotype in HK-2 cells, the rBAT gene was silenced using antisense oligonucleotides complimentary to human rBAT mRNA. The reduced transport activity of cystine was then determined by radiolabeled cystine uptake measurements. RT-PCR and Western blot confirmed the expression of the rBAT gene in HK-2 cells. Considerable transport of the radio labeled cystine was observed in HK-2 cells and was linearly dependent on the incubation time with the amino acid. The cystine transport in rBAT knockdown cells after incubation with antisense oligonucleotides was significantly lower compared to control (0.76 vs. 0.98%; P=0.0008), proving a transient knock-down of the rBAT gene. This study demonstrates the presence of the b(0,+) amino acid transport system in human proximal tubular HK-2 cells when cultured in vitro. Inhibition of this transport system is possible by using antisense technology. A permanent inhibition of the cystine transport, based on our model, would be useful for the development and evaluation gene therapeutic approaches.
Collapse
Affiliation(s)
- Gunnar Wendt-Nordahl
- Department of Urology, University Hospital Mannheim, Theodor-Kutzer-Ufer 1-3, 68135 Mannheim, Germany
| | | | | | | | | | | |
Collapse
|
46
|
Font-Llitjós M, Feliubadaló L, Espino M, Clèries R, Mañas S, Frey IM, Puertas S, Colell G, Palomo S, Aranda J, Visa J, Palacín M, Nunes V. Slc7a9knockout mouse is a good cystinuria model for antilithiasic pharmacological studies. Am J Physiol Renal Physiol 2007; 293:F732-40. [PMID: 17596531 DOI: 10.1152/ajprenal.00121.2007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cystinuria is a hereditary disorder caused by a defect in the apical membrane transport system for cystine and dibasic amino acids in renal proximal tubules and intestine, resulting in recurrent urolithiasis. Mutations in SLC3A1 and SLC7A9 genes, that codify for rBAT/b0,+AT transporter subunits, cause type A and B cystinuria, respectively. In humans, cystinuria treatment is based on the prevention of calculi formation and its dissolution or breakage. Persistent calculi are treated with thiols [i.e., d-penicillamine (DP) and mercaptopropionylglycine (MPG)] for cystine solubilization. We have developed a new protocol with DP to validate our Slc7a9 knockout mouse model for the study of the therapeutic effect of drugs in the treatment of cystine lithiasis. We performed a 5-wk treatment of individually caged lithiasic mutant mice with a previously tested DP dose. To appraise the evolution of lithiasis throughout the treatment a noninvasive indirect method of calculi quantification was developed: calculi mass was quantified by densitometry of X-ray images from cystinuric mice before and after treatment. Urine was collected in metabolic cage experiments to quantify amino acids in DP-treated and nontreated, nonlithiasic mutant mice. We found significant differences between DP-treated and nontreated knockout mice in calculi size and in urinary cystine excretion. Histopathological analysis showed that globally nontreated mutant mice had more severe and diffuse urinary system damage than DP-treated mice. Our results validate the use of this mouse model for testing the efficacy of potential new drugs against cystinuria.
Collapse
Affiliation(s)
- Mariona Font-Llitjós
- Medical and Molecular Genetics Center, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Cystinuria is a monogenic disorder in which there is a transepithelial transport defect of di-basic amino acids, including cystine, ornithine, lysine, and arginine (COLA). This results in diminished reabsorption of these amino acids in both the intestine and renal proximal tubule. This article describes the disorder, reviews the mechanisms of normal COLA renal transport, and summarizes issues related to the disorder, such as the role of mutations, associated diseases, clinical manifestations, therapies, the renal impact, and handling of pediatric patients.
Collapse
|
48
|
Reig N, del Rio C, Casagrande F, Ratera M, Gelpí JL, Torrents D, Henderson PJF, Xie H, Baldwin SA, Zorzano A, Fotiadis D, Palacín M. Functional and Structural Characterization of the First Prokaryotic Member of the L-Amino Acid Transporter (LAT) Family. J Biol Chem 2007; 282:13270-81. [PMID: 17344220 DOI: 10.1074/jbc.m610695200] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have identified YkbA from Bacillus subtilis as a novel member of the L-amino acid transporter (LAT) family of amino acid transporters. The protein is approximately 30% identical in amino acid sequence to the light subunits of human heteromeric amino acid transporters. Purified His-tagged YkbA from Escherichia coli membranes reconstituted in proteoliposomes exhibited sodium-independent, obligatory exchange activity for L-serine and L-threonine and also for aromatic amino acids, albeit with less activity. Thus, we propose that YkbA be renamed SteT (Ser/Thr exchanger transporter). Kinetic analysis supports a sequential mechanism of exchange for SteT. Freeze-fracture analysis of purified, functionally active SteT in proteoliposomes, together with blue native polyacrylamide gel electrophoresis and transmission electron microscopy of detergent-solubilized purified SteT, suggest that the transporter exists in a monomeric form. Freeze-fracture analysis showed spherical particles with a diameter of 7.4 nm. Transmission electron microscopy revealed elliptical particles (diameters 6 x 7 nm) with a distinct central depression. To our knowledge, this is the first functional characterization of a prokaryotic member of the LAT family and the first structural data on an APC (amino acids, polyamines, and choline for organocations) transporter. SteT represents an excellent model to study the molecular architecture of the light subunits of heteromeric amino acid transporters and other APC transporters.
Collapse
Affiliation(s)
- Núria Reig
- Institute for Research in Biomedicine, Barcelona Science Park and Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, E-08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ashman N, Brunini TM, Mann GE, Mendes Ribeiro AC, Yaqoob MM. Increased L-arginine transport via system b0,+ in human proximal tubular cells exposed to albumin. Clin Sci (Lond) 2006; 111:389-99. [PMID: 16928190 DOI: 10.1042/cs20060158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Albumin has complex effects on PTECs (proximal tubular epithelial cells) and is able to stimulate growth or injury depending on its bound moieties. Albumin itself is a mitogen, inducing proliferation through a number of pathways. In PTEC exposed to purified albumin, polyamines are required for entry into the cell cycle and are critical for proliferation. Polyamines are synthesized from L-ornithine (itself derived by the action of arginase on L-arginine), and the transport and availability of L-arginine may thus be important for subsequent polyamine-dependent proliferation. In the present study we investigated radiolabelled cationic amino-acid transport in cultured PTEC exposed to 20 mg/ml ultrapure recombinant human albumin, describing the specific kinetic characteristics of transport and the expression of transporters. L-[3H]Arginine transport capacity in human PTEC is increased after exposure for 24 h to human albumin, mediated by the broad-scope high-affinity system b0,+ and, to a lesser extent, system y+L (but not system y+) transport. Increased transport is associated with increased b0,+-associated transporter expression. Inhibition of phosphoinositide 3-kinase, a key regulator of albumin endocytosis and signalling, inhibited proliferation, but had no effect on the observed increase in transport. PTEC proliferated in response to albumin. L-Lysine, a competitive inhibitor of L-arginine transport, had no effect on albumin-induced proliferation; however, arginine deprivation effectively reversed the albumin-induced proliferation observed. In conclusion, in PTEC exposed to albumin, increased L-arginine transport is mediated by increased transcription and activity of the apical b0,+ transport system. This may make L-arginine available as a substrate for the downstream synthesis of polyamines, but is not critical for cell proliferation.
Collapse
Affiliation(s)
- Neil Ashman
- Department of Experimental Medicine, Critical Care and Nephrology, William Harvey Research Institute, Queen Mary College, University of London, London, UK.
| | | | | | | | | |
Collapse
|
50
|
Fernández E, Jiménez-Vidal M, Calvo M, Zorzano A, Tebar F, Palacín M, Chillarón J. The Structural and Functional Units of Heteromeric Amino Acid Transporters. J Biol Chem 2006; 281:26552-61. [PMID: 16825196 DOI: 10.1074/jbc.m604049200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heteromeric amino acid transporters are composed of a catalytic light subunit and a heavy subunit linked by a disulfide bridge. We analyzed the structural and functional units of systems b0,+ and xC-, formed by the heterodimers b0,+ AT-rBAT and xCT-4F2hc, respectively. Blue Native gel electrophoresis, cross-linking, and fluorescence resonance energy transfer in vivo indicate that system b0,+ is a heterotetramer [b0,+ AT-rBAT]2, whereas xCT-4F2hc seems not to stably or efficiently oligomerize. However, substitution of the heavy subunit 4F2hc for rBAT was sufficient to form a heterotetrameric [xCT-rBAT]2 structure. The functional expression of concatamers of two light subunits (which differ only in their sensitivity to inactivation by a sulfhydryl reagent) suggests that a single heterodimer is the functional unit of systems b0,+ and xC-.
Collapse
Affiliation(s)
- Esperanza Fernández
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|