1
|
Srivastava S, Nakagawa K, He X, Kimura T, Fukutomi T, Miyauchi S, Sakurai H, Anzai N. Identification of the multivalent PDZ protein PDZK1 as a binding partner of sodium-coupled monocarboxylate transporter SMCT1 (SLC5A8) and SMCT2 (SLC5A12). J Physiol Sci 2019; 69:399-408. [PMID: 30604288 PMCID: PMC10717704 DOI: 10.1007/s12576-018-00658-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/18/2018] [Indexed: 12/30/2022]
Abstract
Sodium-coupled monocarboxylate transporters SMCT1 (SLC5A8) and SMCT2 (SLC5A12) mediate the high- and low-affinity transport of lactate in the kidney, but their regulatory mechanism is still unknown. Since these two transporters have the PDZ-motif at their C-terminus, the function of SMCTs may be modulated by a protein-protein interaction. To investigate the binding partner(s) of SMCTs in the kidney, we performed yeast two-hybrid (Y2H) screenings of a human kidney cDNA library with the C-terminus of SMCT1 (SMCT1-CT) and SMCT2 (SMCT2-CT) as bait. PDZK1 was identified as a partner for SMCTs. PDZK1 coexpression in SMCT1-expressing HEK293 cells enhanced their nicotinate transport activity. PDZK1, SMCT1, and URAT1 in vitro assembled into a single tri-molecular complex and their colocalization was confirmed in the renal proximal tubule in vivo by immunohistochemistry. These results indicate that the SMCT1-PDZK1 interaction thus plays an important role in both lactate handling as well as urate reabsorption in the human kidney.
Collapse
Affiliation(s)
- Sunena Srivastava
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Kiyoshi Nakagawa
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| | - Xin He
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Toru Kimura
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Toshiyuki Fukutomi
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Seiji Miyauchi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Toho University, Funabashi, Chiba, Japan
| | - Hiroyuki Sakurai
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan.
| |
Collapse
|
2
|
Kim YK, Nam SA, Yang CW. Applications of kidney organoids derived from human pluripotent stem cells. Korean J Intern Med 2018; 33:649-659. [PMID: 29961307 PMCID: PMC6030416 DOI: 10.3904/kjim.2018.198] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 06/18/2018] [Indexed: 12/11/2022] Open
Abstract
The establishment of protocols to differentiate kidney organoids from human pluripotent stem cells provides potential applications of kidney organoids in regenerative medicine. Modeling of renal diseases, drug screening, nephrotoxicity testing of compounds, and regenerative therapy are attractive applications. Although much progress still remains to be made in the development of kidney organoids, recent advances in clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated system 9 (Cas9) genome editing and three-dimensional bioprinting technologies have contributed to the application of kidney organoids in clinical fields. In this section, we review recent advances in the applications of kidney organoids to kidney disease modelling, drug screening, nephrotoxicity testing, and regenerative therapy.
Collapse
Affiliation(s)
- Yong Kyun Kim
- Cell Death Disease Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sun Ah Nam
- Cell Death Disease Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chul Woo Yang
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Convergent Research Consortium for Immunologic Disease, and Division of Nephrology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Correspondence to Chul Woo Yang, M.D. Convergent Research Consortium for Immunologic Disease and Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-6037 Fax: +82-2-22258-6917 E-mail:
| |
Collapse
|
3
|
Renal Proteomic Responses to Severe Sepsis and Surgical Trauma: Dynamic Analysis of Porcine Tissue Biopsies. Shock 2018; 46:453-64. [PMID: 27070328 DOI: 10.1097/shk.0000000000000613] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although the burden of septic acute kidney injury continues to increase, the molecular pathogenesis remains largely obscure. The aim of this exploratory study was a discovery-driven analysis of dynamic kidney tissue protein expression changes applied for the first time in a classic large mammal model of sepsis. To achieve this goal, analyses of protein expression alterations were performed in serial samples of kidney cortical biopsies (before, 12 and 22 h of sepsis) in mechanically ventilated pigs challenged with continuous infusion of pseudomonas aeruginosa and compared with sham-operated control data. Global protein expression was analyzed using two-dimensional gel electrophoresis and mass spectrometry-based proteomics. Normodynamic sepsis was associated with 43% reduction in glomerular filtration. The exposure to surgical stress per se altered the renal protein expression profile, while sepsis induced distinct and highly dynamic proteome evolution shifting the balance toward cellular distress phenotype. We identified 20 proteins whose expression changes discriminated effects of sepsis from those induced by surgery. The data implicate endoplasmic reticulum stress, oxidative stress, mitochondrial energy metabolism, immune/inflammatory signaling, and tubular transport as major activated pathways. Thus, by coupling the power of sequential tissue proteomics with whole-animal physiological studies, our study helped to establish a first global overview of critical renal proteomic events occurring during surgical trauma and early sepsis in a porcine model. The study supports the notion that multiple potentially subtle and even transient changes in several proteins which are members of key functional interrelated systems appear to play a role in septic acute kidney injury.
Collapse
|
4
|
Abstract
The Lush Science Prize 2016 was awarded to Daniele Zink and Lit-Hsin Loo for the interdisciplinary and collaborative work between their research groups in developing alternative methods for the prediction of nephrotoxicity in humans. The collaboration has led to the establishment of a series of pioneering alternative methods for nephrotoxicity prediction, which includes: predictive gene expression markers based on pro-inflammatory responses; predictive in vitro cellular models based on pluripotent stem cell-derived proximal tubular-like cells; and predictive cellular phenotypic markers based on chromatin and cytoskeletal changes. A high-throughput method was established for chemical testing, which is currently being used to predict the potential human nephrotoxicity of ToxCast compounds in collaboration with the US Environmental Protection Agency. Similar high-throughput imaging-based methodologies are currently being developed and adapted by the Zink and Loo groups, to include other human organs and cell types. The ultimate goal is to develop a portfolio of methods accepted for the accurate prediction of human organ-specific toxicity and the consequent replacement of animal experiments.
Collapse
Affiliation(s)
- Lit-Hsin Loo
- Bioinformatics Institute (BII), Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Daniele Zink
- Institute of Bioengineering and Nanotechnology (IBN), Singapore
| |
Collapse
|
5
|
Chuah JKC, Zink D. Stem cell-derived kidney cells and organoids: Recent breakthroughs and emerging applications. Biotechnol Adv 2016; 35:150-167. [PMID: 28017905 DOI: 10.1016/j.biotechadv.2016.12.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/12/2016] [Accepted: 12/17/2016] [Indexed: 02/09/2023]
Abstract
The global rise in the numbers of kidney patients and the shortage in transplantable organs have led to an increasing interest in kidney-specific regenerative therapies, renal disease modelling and bioartificial kidneys. Sources for large quantities of high-quality renal cells and tissues would be required, also for applications in in vitro platforms for compound safety and efficacy screening. Stem cell-based approaches for the generation of renal-like cells and tissues would be most attractive, but such methods were not available until recently. This situation has drastically changed since 2013, and various protocols for the generation of renal-like cells and precursors from pluripotent stem cells (PSC) have been established. The most recent breakthroughs were related to the establishment of various protocols for the generation of PSC-derived kidney organoids. In combination with recent advances in genome editing, bioprinting and the establishment of predictive renal screening platforms this results in exciting new possibilities. This review will give a comprehensive overview over current PSC-based protocols for the generation of renal-like cells, precursors and organoids, and their current and potential applications in regenerative medicine, compound screening, disease modelling and bioartificial organs.
Collapse
Affiliation(s)
- Jacqueline Kai Chin Chuah
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Daniele Zink
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| |
Collapse
|
6
|
Kawano T, Ryuda M, Matsumoto H, Ochiai M, Oda Y, Tanimura T, Csikos G, Moriya M, Hayakawa Y. Function of desiccate in gustatory sensilla of drosophila melanogaster. Sci Rep 2015; 5:17195. [PMID: 26610608 PMCID: PMC4661605 DOI: 10.1038/srep17195] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/27/2015] [Indexed: 12/02/2022] Open
Abstract
Desiccate (Desi), initially discovered as a gene expressing in the epidermis of Drosophila larvae for protection from desiccation stress, was recently found to be robustly expressed in the adult labellum; however, the function, as well as precise expression sites, was unknown. Here, we found that Desi is expressed in two different types of non-neuronal cells of the labellum, the epidermis and thecogen accessory cells. Labellar Desi expression was significantly elevated under arid conditions, accompanied by an increase in water ingestion by adults. Desi overexpression also promoted water ingestion. In contrast, a knockdown of Desi expression reduced feeding as well as water ingestion due to a drastic decrease in the gustatory sensillar sensitivity for all tested tastants. These results indicate that Desi helps protect insects from desiccation damage by not only preventing dehydration through the integument but also accelerating water ingestion via elevated taste sensitivities of the sensilla.
Collapse
Affiliation(s)
- Takeshi Kawano
- Department of Applied Biological Sciences, Saga University, Saga 840-8502, Japan
| | - Masasuke Ryuda
- The Analytical Research Center for Experimental Sciences of Saga University, Saga 840-8502, Japan
| | - Hitoshi Matsumoto
- Department of Applied Biological Sciences, Saga University, Saga 840-8502, Japan
| | - Masanori Ochiai
- Institute of Low Temperature Science, Hokkaido University, Sapporo 060-0819, Japan
| | - Yasunori Oda
- Department of Applied Biological Sciences, Saga University, Saga 840-8502, Japan
| | - Teiichi Tanimura
- Department of Biology, Graduate School of Sciences, Kyushu University, Hakozaki, Fukuoka 812-8581, Japan
| | - Gyorge Csikos
- Department of Anatomy, Cell and Molecular Biology, Eotvos Lorand University, Budapest, Hungary, H-1117
| | - Megumi Moriya
- Institute of Low Temperature Science, Hokkaido University, Sapporo 060-0819, Japan
| | - Yoichi Hayakawa
- Department of Applied Biological Sciences, Saga University, Saga 840-8502, Japan
| |
Collapse
|
7
|
Ko B, Bergsland K, Gillen DL, Evan AP, Clark DL, Baylock J, Coe FL, Worcester EM. Sex differences in proximal and distal nephron function contribute to the mechanism of idiopathic hypercalcuria in calcium stone formers. Am J Physiol Regul Integr Comp Physiol 2015; 309:R85-92. [PMID: 25947170 DOI: 10.1152/ajpregu.00071.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 04/30/2015] [Indexed: 11/22/2022]
Abstract
Idiopathic hypercalciuria (IH) is a common familial trait among patients with calcium nephrolithiasis. Previously, we have demonstrated that hypercalciuria is primarily due to reduced renal proximal and distal tubule calcium reabsorption. Here, using measurements of the clearances of sodium, calcium, and endogenous lithium taken from the General Clinical Research Center, we test the hypothesis that patterns of segmental nephron tubule calcium reabsorption differ between the sexes in IH and normal subjects. When the sexes are compared, we reconfirm the reduced proximal and distal calcium reabsorption. In IH women, distal nephron calcium reabsorption is decreased compared to normal women. In IH men, proximal tubule calcium reabsorption falls significantly, with a more modest reduction in distal calcium reabsorption compared to normal men. Additionally, we demonstrate that male IH patients have lower systolic blood pressures than normal males. We conclude that women and men differ in the way they produce the hypercalciuria of IH, with females reducing distal reabsorption and males primarily reducing proximal tubule function.
Collapse
Affiliation(s)
- Benjamin Ko
- Department of Medicine, University of Chicago School of Medicine, Chicago, Illinois;
| | - Kristin Bergsland
- Department of Medicine, University of Chicago School of Medicine, Chicago, Illinois
| | - Daniel L Gillen
- Department of Statistics, University of California, Irvine, California; and
| | - Andrew P Evan
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Daniel L Clark
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jaime Baylock
- Department of Medicine, University of Chicago School of Medicine, Chicago, Illinois
| | - Fredric L Coe
- Department of Medicine, University of Chicago School of Medicine, Chicago, Illinois
| | - Elaine M Worcester
- Department of Medicine, University of Chicago School of Medicine, Chicago, Illinois
| |
Collapse
|
8
|
Hering-Smith KS, Mao W, Schiro FR, Coleman-Barnett J, Pajor AM, Hamm LL. Localization of the calcium-regulated citrate transport process in proximal tubule cells. Urolithiasis 2014; 42:209-19. [PMID: 24652587 DOI: 10.1007/s00240-014-0653-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 02/25/2014] [Indexed: 11/26/2022]
Abstract
Urinary citrate is an important inhibitor of calcium-stone formation. Most of the citrate reabsorption in the proximal tubule is thought to occur via a dicarboxylate transporter NaDC1 located in the apical membrane. OK cells, an established opossum kidney proximal tubule cell line, transport citrate but the characteristics change with extracellular calcium such that low calcium solutions stimulate total citrate transport as well as increase the apparent affinity for transport. The present studies address several fundamental properties of this novel process: the polarity of the transport process, the location of the calcium-sensitivity and whether NaDC1 is present in OK cells. OK cells grown on permeable supports exhibited apical >basolateral citrate transport. Apical transport of both citrate and succinate was sensitive to extracellular calcium whereas basolateral transport was not. Apical calcium, rather than basolateral, was the predominant determinant of changes in transport. Also 2,3-dimethylsuccinate, previously identified as an inhibitor of basolateral dicarboxylate transport, inhibited apical citrate uptake. Although the calcium-sensitive transport process in OK cells is functionally not typical NaDC1, NaDC1 is present in OK cells by Western blot and PCR. By immunolocalization studies, NaDC1 was predominantly located in discrete apical membrane or subapical areas. However, by biotinylation, apical NaDC1 decreases in the apical membrane with lowering calcium. In sum, OK cells express a calcium-sensitive/regulated dicarboxylate process at the apical membrane which responds to variations in apical calcium. Despite the functional differences of this process compared to NaDC1, NaDC1 is present in these cells, but predominantly in subapical vesicles.
Collapse
Affiliation(s)
- Kathleen S Hering-Smith
- Research Service, Southeastern Louisiana Veterans Health Care System (SLVHCS), New Orleans, LA, 70161, USA,
| | | | | | | | | | | |
Collapse
|
9
|
Park J, Kwak JO, Riederer B, Seidler U, Cole SPC, Lee HJ, Lee MG. Na⁺/H⁺ exchanger regulatory factor 3 is critical for multidrug resistance protein 4-mediated drug efflux in the kidney. J Am Soc Nephrol 2014; 25:726-36. [PMID: 24436471 DOI: 10.1681/asn.2013040438] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Na(+)/H(+) exchanger regulatory factor 3 (NHERF3) is a PSD-95/discs large/ZO-1 (PDZ)-based adaptor protein that regulates several membrane-transporting proteins in epithelia. However, the in vivo physiologic role of NHERF3 in transepithelial transport remains poorly understood. Multidrug resistance protein 4 (MRP4) is an ATP binding cassette transporter that mediates the efflux of organic molecules, such as nucleoside analogs, in the gastrointestinal and renal epithelia. Here, we report that Nherf3 knockout (Nherf3(-/-)) mice exhibit profound reductions in Mrp4 expression and Mrp4-mediated drug transport in the kidney. A search for the binding partners of the COOH-terminal PDZ binding motif of MRP4 among several epithelial PDZ proteins indicated that MRP4 associated most strongly with NHERF3. When expressed in HEK293 cells, NHERF3 increased membrane expression of MRP4 by reducing internalization of cell surface MRP4 and consequently, augmented MRP4-mediated efflux of adefovir, a nucleoside-based antiviral agent and well known substrate of MRP4. Examination of wild-type and Nherf3(-/-) mice revealed that Nherf3 is most abundantly expressed in the kidney and has a prominent role in modulating Mrp4 levels. Deletion of Nherf3 in mice caused a profound reduction in Mrp4 expression at the apical membrane of renal proximal tubules and evoked a significant increase in the plasma and kidney concentrations of adefovir, with a corresponding decrease in the systemic clearance of this drug. These results suggest that NHERF3 is a key regulator of organic transport in the kidney, particularly MRP4-mediated clearance of drug molecules.
Collapse
Affiliation(s)
- Joonhee Park
- Department of Pharmacology and Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Organic anions and cations (OAs and OCs, respectively) comprise an extraordinarily diverse array of compounds of physiological, pharmacological, and toxicological importance. The kidney, primarily the renal proximal tubule, plays a critical role in regulating the plasma concentrations of these organic electrolytes and in clearing the body of potentially toxic xenobiotics agents, a process that involves active, transepithelial secretion. This transepithelial transport involves separate entry and exit steps at the basolateral and luminal aspects of renal tubular cells. Basolateral and luminal OA and OC transport reflects the concerted activity of a suite of separate proteins arranged in parallel in each pole of proximal tubule cells. The cloning of multiple members of several distinct transport families, the subsequent characterization of their activity, and their subcellular localization within distinct regions of the kidney, now allows the development of models describing the molecular basis of the renal secretion of OAs and OCs. New information on naturally occurring genetic variation of many of these processes provides insight into the basis of observed variability of drug efficacy and unwanted drug-drug interactions in human populations. The present review examines recent work on these issues.
Collapse
Affiliation(s)
- Ryan M Pelis
- Novartis Pharmaceuticals Corp., Translational Sciences, East Hanover, New Jersey, USA
| | | |
Collapse
|
11
|
Abstract
Hyperuricemia is associated with an increased risk of developing gout. This increases with the degree and duration of hyperuricemia. Gout can be managed by dietary modification and pharmacologic urate-lowering therapies. The recent identification of the renal apical urate/anion exchanger URAT1 (SLC22A12) and several membrane proteins relevant to the transport of urate play an important role in gaining a better understanding of the mode of action of many drugs used to treat gout. As described in this review, therapeutics designed to modify URAT1 transport activities might be useful in treating pathologies associated with hyperuricemia such as gout and urolithiasis. Continuing studies into the urate transportsome hold promise for the development of new, more effective therapeutics for hyperuricemia.
Collapse
Affiliation(s)
- Naohiko Anzai
- Kyorin University School of Medicine, Department of Pharmacology and Toxicology, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan +81 422 47 5511 (ext 3692) ; +81 422 79 1321 ;
| | | |
Collapse
|
12
|
Fujimoto T, Shirasawa S. Identification of KRAP-expressing cells and the functional relevance of KRAP to the subcellular localization of IP3R in the stomach and kidney. Int J Mol Med 2012; 30:1287-93. [PMID: 22992961 PMCID: PMC4042864 DOI: 10.3892/ijmm.2012.1126] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 08/20/2012] [Indexed: 12/31/2022] Open
Abstract
KRAS-induced actin-interacting protein (KRAP), originally identified as
one of the deregulated genes expressed in colorectal cancer, participates under
physiological conditions in the regulation of systemic energy homeostasis and of the
exocrine system. We have recently found that KRAP is a molecule associated with inositol
1,4,5-trisphosphate receptor (IP3R) and is critical for the proper subcellular
localization of IP3R in the liver and the pancreas. However, the expression of
KRAP and its precise function in other tissues remain elusive. In this study, we aimed to
identify the KRAP-expressing cells in mouse stomach and kidneys and to examine the
relevance of KRAP expression in the regulation of IP3R localization in these
tissues. In the stomach, double immunohistochemical staining for KRAP and IP3R
demonstrated that KRAP was expressed along with the apical regions in the mucous cells and
the chief cells, and IP3R3 was dominantly co-localized with KRAP in these
cells. Furthermore, IP3R2 was also co-localized with IP3R3 in the
chief cells. It is of note that the proper localization of IP3R3 and
IP3R2 in the chief cells and of IP3R3 in the mucous cells were
significantly abrogated in KRAP-deficient mice. In the kidneys, KRAP was
expressed in both the apical and the basal regions of the proximal tubular cells.
Intriguingly, KRAP deficiency abrogated the localization of
IP3R1 in the proximal tubular cells. Finally, co-immunoprecipitation study in
the stomachs and the kidneys validated the physical association of KRAP with
IP3Rs. These findings demonstrate that KRAP physically associates with
IP3Rs and regulates the proper localization of IP3Rs in the mucous
cells and the chief cells of the stomach and in the proximal tubular cells of the
kidneys.
Collapse
Affiliation(s)
- Takahiro Fujimoto
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | | |
Collapse
|
13
|
Tasnim F, Deng R, Hu M, Liour S, Li Y, Ni M, Ying JY, Zink D. Achievements and challenges in bioartificial kidney development. FIBROGENESIS & TISSUE REPAIR 2010; 3:14. [PMID: 20698955 PMCID: PMC2925816 DOI: 10.1186/1755-1536-3-14] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 08/10/2010] [Indexed: 12/23/2022]
Abstract
Bioartificial kidneys (BAKs) combine a conventional hemofilter in series with a bioreactor unit containing renal epithelial cells. The epithelial cells derived from the renal tubule should provide transport, metabolic, endocrinologic and immunomodulatory functions. Currently, primary human renal proximal tubule cells are most relevant for clinical applications. However, the use of human primary cells is associated with many obstacles, and the development of alternatives and an unlimited cell source is one of the most urgent challenges. BAKs have been applied in Phase I/II and Phase II clinical trials for the treatment of critically ill patients with acute renal failure. Significant effects on cytokine concentrations and long-term survival were observed. A subsequent Phase IIb clinical trial was discontinued after an interim analysis, and these results showed that further intense research on BAK-based therapies for acute renal failure was required. Development of BAK-based therapies for the treatment of patients suffering from end-stage renal disease is even more challenging, and related problems and research approaches are discussed herein, along with the development of mobile, portable, wearable and implantable devices.
Collapse
Affiliation(s)
- Farah Tasnim
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Amatschek S, Haller M, Oberbauer R. Renal phosphate handling in human--what can we learn from hereditary hypophosphataemias? Eur J Clin Invest 2010; 40:552-60. [PMID: 20412291 DOI: 10.1111/j.1365-2362.2010.02286.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Renal reabsorption of inorganic phosphate is critical for the maintenance of phosphate homeostasis. The sodium dependent phosphate cotransporters NaPi-IIa and NaPi-IIc have been identified to fulfill this task at the brush border membrane of proximal tubule cells. Various factors including dietary phosphate intake, parathyroid hormone, or the so called phosphatonins such as FGF23 have been shown to regulate activity of these transporters. DESIGN This review seeks to give an update on our current knowledge about regulatory mechanisms involved in human renal phosphate reabsorption. RESULTS Recently, an increasing number of genes have been identified that are directly associated with inherited phosphate wasting disorders (Klotho, PHEX, DMP1 and NHERF1). Several of these genes are predominantly expressed by osteocytes and osteoclasts in the bone suggesting indispensable signalling pathways between kidneys and the skeleton. CONCLUSION In this review, the affected gene products in these inherited hypophosphataemias and their contribution to phosphate homeostasis are discussed.
Collapse
|
15
|
Unger S, Tausche AK, Kopprasch S, Bornstein SR, Aringer M, Grässler J. [Molecular basis of primary renal hyperuricemia : role of the human urate transporter hURAT1]. Z Rheumatol 2008; 66:556, 58-61. [PMID: 17891408 DOI: 10.1007/s00393-007-0208-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In highly industrialized countries hyperuricemia is one of the most common metabolic disorders. High uric acid blood levels may lead to the manifestation of gout owing to the precipitation of urate crystals in connective tissue, the skeletal system and kidneys. A primary reduction of renal uric acid excretion can be detected in more than 90% of all cases of hyperuricemia. Despite the identification of several uric acid transporting proteins their pathogenetic role for the induction of primary reduced renal uric acid excretion has not yet been verified. As a result of a case-control study on individuals with normal and reduced renal uric acid excretion, an association of polymorphisms in the human urate transporter 1 gene (hURAT1) with primary reduced urate excretion has been demonstrated for the first time. The hURAT1 gene is an organic anion transporter (SLC22A12), which is preferentially expressed in the apical membrane of proximal renal tubule cells. Functioning as an antiporter, hURAT1 mediates the uptake of urate from the lumen into proximal tubule cells in exchange for organic and inorganic anions. Loss-of-function mutations in the hURAT1 gene are a cause of hereditary renal hypouricemia. The precisely regulated hURAT1 is a candidate gene for hyperuricemia and an important target for the development and optimization of new diagnostic approaches and pharmacological interventions of primary reduced renal uric acid excretion.
Collapse
Affiliation(s)
- S Unger
- Bereich Pathologische Biochemie, Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Deutschland
| | | | | | | | | | | |
Collapse
|
16
|
Truong DM, Kaler G, Khandelwal A, Swaan PW, Nigam SK. Multi-level analysis of organic anion transporters 1, 3, and 6 reveals major differences in structural determinants of antiviral discrimination. J Biol Chem 2008; 283:8654-63. [PMID: 18174163 PMCID: PMC2417182 DOI: 10.1074/jbc.m708615200] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 12/04/2007] [Indexed: 11/06/2022] Open
Abstract
Long-term exposure to antivirals is associated with serious cellular toxicity to the kidney and other tissues. Organic anion transporters (OATs) are believed to mediate the cellular uptake, and hence cytotoxicity, of many antivirals. However, a systematic in vitro and ex vivo analysis of interactions between these compounds with various OAT isoforms has been lacking. To characterize substrate interactions with mOat1, mOat3, and mOat6, a fluorescence-based competition assay in Xenopus oocytes as well as wild-type and knock-out whole embryonic kidney (WEK) organ culture systems was developed using 6-carboxyfluorescein, 5-carboxyfluorescein, and fluorescein. Of nine common antiviral drugs assessed in oocytes, many manifested higher affinity for SLC22a6 (mOat1), originally identified as NKT (e.g. adefovir and cidofovir), two (ddC and ddI) manifested significantly higher affinity for mOat3, while mOat6 had comparatively low but measurable affinity for certain antivirals. A live organ staining approach combined with fluorescent uptake in WEK cultures allowed the visualization of OAT-mediated uptake ex vivo into developing proximal tubule-like structures, as well as quantification of substrate interactions of individual OAT isoforms. In general, antiviral specificity of SLC22a6 (Oat1) (in Oat3(-/-) WEK culture) and SLC22a8 (Oat3) (in Oat1(-/-) WEK culture) was consistent with the Xenopus oocyte data. The combined observations suggest SLC22a8 (Oat3) is the major transporter interacting with ddC and ddI. Finally, quantitative structure-activity relationship analysis of the nine antivirals' physicochemical descriptors with their OAT affinity indicates that antiviral preferences of mOat1 are explained by high polar surface areas (e.g. phosphate groups), whereas mOat3 prefers hydrogen bond acceptors (e.g. amines, ketones) and low rotatable bond numbers. In contrast, hydrogen bond donors (e.g. amides, alcohols) diminish binding to mOat6. This suggests that, despite sharing close overall sequence homology, Oat1, Oat3, and Oat6 have signficantly different binding pockets. Taken together, the data provide a basis for understanding potential drug interactions in combination antiviral therapy, as well as suggesting structural mdifications for drug design, especially in the context of targeting toward or away from specific tissues.
Collapse
Affiliation(s)
- David M Truong
- Department of Pediatrics, University of California at San Diego, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
17
|
Sabolić I, Asif AR, Budach WE, Wanke C, Bahn A, Burckhardt G. Gender differences in kidney function. Pflugers Arch 2007; 455:397-429. [PMID: 17638010 DOI: 10.1007/s00424-007-0308-1] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 06/12/2007] [Accepted: 06/14/2007] [Indexed: 01/04/2023]
Abstract
Sex hormones influence the development of female (F) and male (M) specific traits and primarily affect the structure and function of gender-specific organs. Recent studies also indicated their important roles in regulating structure and/or function of nearly every tissue and organ in the mammalian body, including the kidneys, causing gender differences in a variety of characteristics. Clinical observations in humans and studies in experimental animals in vivo and in models in vitro have shown that renal structure and functions under various physiological, pharmacological, and toxicological conditions are different in M and F, and that these differences may be related to the sex-hormone-regulated expression and action of transporters in the apical and basolateral membrane of nephron epithelial cells. In this review we have collected published data on gender differences in renal functions, transporters and other related parameters, and present our own microarray data on messenger RNA expression for various transporters in the kidney cortex of M and F rats. With these data we would like to emphasize the importance of sex hormones in regulation of a variety of renal transport functions and to initiate further studies of gender-related differences in kidney structure and functions, which would enable us to better understand occurrence and development of various renal diseases, pharmacotherapy, and drug-induced nephrotoxicity in humans and animals.
Collapse
Affiliation(s)
- Ivan Sabolić
- Molecular Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | | | | | | | | | | |
Collapse
|
18
|
Ho HTB, Ko BCB, Cheung AKH, Lam AKM, Tam S, Chung SK, Chung SSM. Generation and characterization of sodium-dicarboxylate cotransporter-deficient mice. Kidney Int 2007; 72:63-71. [PMID: 17410095 DOI: 10.1038/sj.ki.5002258] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The sodium-dependent dicarboxylate cotransporter (NaDC1) has a proposed function of reabsorbing various Krebs cycle intermediates in the kidney and the small intestine. Since Krebs cycle intermediates have been suggested to be important for renal cell survival and recovery after hypoxia and reoxygenation, the transporter may play a role in the recovery of the kidney. Additionally, mutations in the transporter homolog in Drosophila led to fly longevity which was thought to be similar to that induced by caloric restriction (CR). To clarify the role of the sodium dicarboxylate cotransporter in vivo we generated cotransporter-deficient mice. These knockout mice excreted significantly higher amounts of various Krebs cycle intermediates in their urine; thus confirming the proposed function to reabsorb these metabolic intermediates in the kidney. No other phenotypic change was identified in these mice, however. Transporter deficiency did not affect renal function under normal physiological conditions, nor did it have an effect on renal damage and recovery from ischemic injury. Additionally, the absence of the transporter did not lead to metabolic or physiological changes associated with CR. Our results suggest that although the sodium dicarboxylate cotransporter is involved in regulating levels of various Krebs cycle intermediates in the kidney, impaired uptake of these intermediates does not significantly affect renal function under normal or ischemic stress.
Collapse
Affiliation(s)
- H T B Ho
- Department of Physiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Rizwan AN, Burckhardt G. Organic anion transporters of the SLC22 family: biopharmaceutical, physiological, and pathological roles. Pharm Res 2007; 24:450-70. [PMID: 17245646 DOI: 10.1007/s11095-006-9181-4] [Citation(s) in RCA: 195] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Accepted: 10/19/2006] [Indexed: 02/08/2023]
Abstract
The human organic anion transporters OAT1, OAT2, OAT3, OAT4 and URAT1 belong to a family of poly-specific transporters mainly located in kidneys. Selected OATs occur also in liver, placenta, and brain. OATs interact with endogenous metabolic end products such as urate and acidic neutrotransmitter metabolites, as well as with a multitude of widely used drugs, including antibiotics, antihypertensives, antivirals, anti-inflammatory drugs, diuretics and uricosurics. Thereby, OATs play an important role in renal drug elimination and have an impact on pharmacokinetics. In this review we focus on the interaction of human OATs with drugs. We report the affinities of human OATs for drug classes and compare the putative importance of individual OATs for renal drug excretion. The role of OATs as sites of drug-drug interaction and mediators cell toxicity, their gender-dependent regulation in health and diseased states, and the possible impact of single nucleotide polymorphisms are also dealt with.
Collapse
Affiliation(s)
- Ahsan N Rizwan
- Abteilung Vegetative Physiologie und Pathophysiologie, Bereich Humanmedizin, Georg-August-Universität Göttingen, Humboldtallee 23, 37073, Göttingen, Germany
| | | |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW This review focuses on recent progress in the understanding of various aspects of renal transport of urate. RECENT FINDINGS Since the molecular cloning of the renal apical urate/anion exchanger URAT1 (SLC22A12), several membrane proteins relevant to the transport of urate have been identified. The molecular identification of two sodium-coupled monocarboxylate transporters, SMCT1(SLC5A8) and SMCT2(SLC5A12), and the emerging role of PDZ (PSD-95, DglA, and ZO-1) scaffold for renal apical transporters have led to a new concept of renal urate transport: urate-transporting multimolecular complex, or 'urate transportsome', that may form an ultimate functional unit including the sodium-coupled urate transport system by linking URAT1 and sodium-coupled monocarboxylate transporters or the coordinated apical urate uptake system by balancing reabsorptive (URAT1) and efflux (NPT1/OATv1 and MRP4) transporters. In addition, genetic variations of the URAT1 gene are associated not only with idiopathic renal hypouricemia but also with reduced renal urate excretion. SUMMARY Although our knowledge of renal urate handling has been increased by the molecular identification of urate transport proteins and by results of genetic studies on patients with serum urate disorders, current evidence is insufficient to fully understand the precise mechanism governing the bi-directional transport of urate. Further studies are still necessary.
Collapse
Affiliation(s)
- Naohiko Anzai
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Shinkawa, Mitaka-shi, Tokyo, Japan
| | | | | |
Collapse
|
21
|
Durand E, Chaumet-Riffaud P, Prigent A. Is it possible to predict renal function in small animals using a multi-pinhole SPECT system? Eur J Nucl Med Mol Imaging 2007; 34:606. [PMID: 17221183 DOI: 10.1007/s00259-006-0336-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
22
|
Nilwarangkoon S, Anzai N, Shiraya K, Yu E, Islam R, Cha SH, Onozato ML, Miura D, Jutabha P, Tojo A, Kanai Y, Endou H. Role of Mouse Organic Anion Transporter 3 (mOat3) as a Basolateral Prostaglandin E2 Transport Pathway. J Pharmacol Sci 2007; 103:48-55. [PMID: 17220594 DOI: 10.1254/jphs.fp0060816] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Renal organic anion transporters play an important role in the handling of a number of endogenous and exogenous anionic substances in the kidney. In this study, we investigated prostaglandin E(2) (PGE(2)) transport properties and intrarenal localization of mouse organic anion transporter 3 (mOat3). When expressed in Xenopus oocytes, mOat3 mediated the time- and concentration-dependent transport of PGE(2) (K(m): 1.48 microM). PGE(2) transport mediated by mOat3 was trans-stimulated by intracellular glutarate injected into the oocytes. PGE(2) efflux via mOat3 was also trans-stimulated by extracellular glutarate. Thus, mOat3 was shown to mediate the bidirectional transport of PGE(2), partly coupled to the dicarboxylate exchange mechanism. Immunohistochemical study revealed that mOat3 protein was localized at the basolateral membrane of renal proximal and distal tubules. Furthermore, diffuse expression of mOat3, including expression in the basolateral membrane in macula densa (MD) cells, was observed. These results indicate that mOat3 plays an important role as a basolateral transport pathway of PGE(2) in the distal nephron including MD cells that may constitute one of the indispensable steps for renin release and regulation of the tubuloglomerular feedback mechanism.
Collapse
Affiliation(s)
- Sirinun Nilwarangkoon
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Sadakata T, Washida M, Morita N, Furuichi T. Tissue distribution of Ca2+-dependent activator protein for secretion family members CAPS1 and CAPS2 in mice. J Histochem Cytochem 2006; 55:301-11. [PMID: 17164411 DOI: 10.1369/jhc.6a7033.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The family of Ca2+-dependent activator proteins for secretion (CAPS) is involved in dense-core vesicle exocytosis. CAPS1/CADPS1 and CAPS2/CADPS2 have been identified in mammals. CAPS1 regulates catecholamine release from neuroendocrine cells, whereas CAPS2 is involved in the release of brain-derived neurotrophic factor and neurotrophin-3 from cerebellar granule cells. CAPS1 and CAPS2 are predominantly expressed in brain. Here we show the immunohistochemical localization of the CAPS family proteins in various mouse tissues. In the pituitary gland, CAPS1 and CAPS2 were localized to the pars nervosa and the pars intermedia, respectively. In non-neural tissues, CAPS1 was observed in the islets of Langerhans, minor cell types of the spleen and stomach, and medullary cells of the adrenal gland, whereas CAPS2 was present in bronchial epithelial cells, thyroid parafollicular cells, chief cells of the stomach, ductal epithelium of the salivary gland, kidney proximal tubules, and minor cell types of the thymus, spleen, and colon. These results suggest that secretion from distinct cell types in various tissues involves either or both members of the CAPS family.
Collapse
Affiliation(s)
- Tetsushi Sadakata
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | | | | | | |
Collapse
|
24
|
Lam HD, Lemay AM, Kelly J, Hill CE. Loss of Kv and MaxiK currents associated with increased MRP1 expression in small cell lung carcinoma. J Cell Physiol 2006; 209:535-41. [PMID: 16883578 DOI: 10.1002/jcp.20761] [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: 01/11/2023]
Abstract
Regulatory volume decrease and exocrine secretion studies suggest a functional relationship between K+ and organic anion efflux. To test the hypothesis that the expression of K+ channels and MRP1 is reciprocally related, we employed the patch clamp and RT-PCR techniques on weakly (H69) and strongly MRP1-expressing (H69AR) small cell lung cancer cells. H69AR cells do not express the time- and voltage-dependent delayed rectifying K+ current (Kv) reported earlier in H69 cells and confirmed here. About 80% of the Kv current in H69 cells inactivated at 0 mV, allowing us to identify other K+ currents present in these cells. Whole-cell currents from cells dialyzed and bathed in K-gluconate as the major ions exhibited inward rectification in both cell types. Inwardly rectifying (Kir) currents in both H69 and H69AR cells showed time-dependent activation and slow inactivation at large negative potentials. H69 cells also express a threefold larger Ca2+ -stimulated K+ -selective and iberiotoxin-sensitive current relative to H69AR cells. In excised inside-out patches exposed to 145 mM symmetrical K+ solutions, H69 cells expressed a voltage- and Ca2+ -sensitive large conductance (128 +/- 5 pS) K+ channel (MaxiK). MaxiK-like currents were not observed at the whole-cell or single-channel level in H69AR cells. RT-PCR identified MaxiKalpha transcripts in H69 but not H69AR cells. These results indicate that two K+ currents (MaxiK and Kv) and the organic anion transporter MRP1 are reciprocally expressed in H69 and H69AR cells.
Collapse
Affiliation(s)
- Hung D Lam
- GI Diseases Research Unit, Hotel Dieu Hospital and Queen's University, Kingston, Ontario, Canada
| | | | | | | |
Collapse
|
25
|
Abstract
Organic anion transporters (OATs) play an essential role in the elimination of numerous endogenous and exogenous organic anions from the body. The renal OATs contribute to the excretion of many drugs and their metabolites that are important in clinical medicine. Several families of multispecific organic anion and cation transporters, including OAT family transporters, have recently been identified by molecular cloning. The OAT family consists of six isoforms (OAT1 - 4, URAT1, and rodent Oat5) and they are all expressed in the kidney, while some are also expressed in the liver, brain, and placenta. The OAT family represents mainly the renal secretory and reabsorptive pathway for organic anions and is also involved in the distribution of organic anions in the body, drug-drug interactions, and toxicity of anionic substances such as nephrotoxic drugs and uremic toxins. In this review, current knowledge of and recent progress in the understanding of several aspects of OAT family members are discussed.
Collapse
Affiliation(s)
- Naohiko Anzai
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Tokyo, Japan
| | | | | |
Collapse
|
26
|
Noshiro R, Anzai N, Sakata T, Miyazaki H, Terada T, Shin HJ, He X, Miura D, Inui K, Kanai Y, Endou H. The PDZ domain protein PDZK1 interacts with human peptide transporter PEPT2 and enhances its transport activity. Kidney Int 2006; 70:275-82. [PMID: 16738539 DOI: 10.1038/sj.ki.5001522] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The proton-coupled peptide transporter PEPT2 (SLC15A2) mediates the high-affinity low-capacity transport of small peptides as well as various oral peptide-like drugs in the kidney. In contrast to its well-characterized transport properties, there is less information available on its regulatory mechanism, although the interaction of PEPT2 to the PDZ (PSD-95, DglA, and ZO-1)-domain protein PDZK1 has been preliminarily reported. To examine whether PDZK1 is a physiological partner of PEPT2 in kidneys, we started from a yeast two-hybrid screen of a human kidney cDNA library with the C-terminus of PEPT2 (PEPT2 C-terminus (PEPT2-CT)) as bait. We could identify PDZK1 as one of the positive clones. This interaction requires the PDZ motif of PEPT2-CT detected by a yeast two-hybrid assay, in vitro binding assay and co-immunoprecipitation. The binding affinities of second and third PDZ domains of PDZK1 to PEPT2-CT were measured by surface plasmon resonance. Co-immunoprecipitation using human kidney membrane fraction and localization of PEPT2 in renal apical proximal tubules revealed the physiological meaning of this interaction in kidneys. Furthermore, we clarified the mechanism of enhanced glycylsarcosine (Gly-Sar) transport activity in PEPT2-expressing HEK293 cells after the PDZK1 coexpression. This augmentation was accompanied by a significant increase in the V(max) of Gly-Sar transport via PEPT2 and it was also associated with the increased surface expression level of PEPT2. These results indicate that the PEPT2-PDZK1 interaction thus plays a physiologically important role in both oligopeptide handling as well as peptide-like drug transport in the human kidney.
Collapse
Affiliation(s)
- R Noshiro
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sugiura T, Kato Y, Tsuji A. Role of SLC xenobiotic transporters and their regulatory mechanisms PDZ proteins in drug delivery and disposition. J Control Release 2006; 116:238-46. [PMID: 16876283 DOI: 10.1016/j.jconrel.2006.06.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Accepted: 06/06/2006] [Indexed: 01/09/2023]
Abstract
Various types of xenobiotic (or drug) transporters have been recently identified to play important roles as barriers against toxic compounds and influx pumps to take up nutrients into the body. Since those xenobiotic transporters generally have wide range of recognition specificity and accept various types of compounds as substrates, localization and functional expression of such transporters could be one of the critical factors that affect the disposition and subsequent biological activity of therapeutic agents. Identification and characterization of drug transporters have given us a scientific basis for understanding drug delivery and disposition, as well as the molecular mechanisms of drug interaction and inter-individual/inter-species differences. To precisely understand pharmacological roles of the transporters in the body, it is also important to clarify molecular mechanisms involved in regulation of the transporters. As a first step to clarify the regulatory mechanisms that govern cell-surface expression and/or function of these transporters, recent researches have focused on PDZ (PSD-95/Discs-large/ZO-1) binding motif localized on carboxylic terminus of several types of xenobiotic transporters. Most of the transporters showing direct interaction potential with the PDZ domain-containing proteins are expressed on apical membranes in epithelial cells of kidney and/or small intestine, implying that such protein-protein interaction may play a role in apical localization of the transporters. In this mini-review article, we summarize importance of transporters and their regulatory mechanisms in drug delivery and disposition, focusing on several aspects of transporter-mediated drug targeting.
Collapse
Affiliation(s)
- Tomoko Sugiura
- Division of Pharmaceutical Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | | | | |
Collapse
|
28
|
Brandoni A, Anzai N, Kanai Y, Endou H, Torres AM. Renal elimination of p-aminohippurate (PAH) in response to three days of biliary obstruction in the rat. The role of OAT1 and OAT3. Biochim Biophys Acta Mol Basis Dis 2006; 1762:673-82. [PMID: 16844357 DOI: 10.1016/j.bbadis.2006.05.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Revised: 05/24/2006] [Accepted: 05/27/2006] [Indexed: 12/18/2022]
Abstract
Pharmacokinetic studies of the drugs administered to subjects with mechanical cholestasis are scarce. The purpose of the present study was to examine the effects of bile duct ligation of 3 days (peak of elevation of serum bile acids and bilirubin) on the systemic and renal PAH clearance and on the expression of cortical renal OAT1 and OAT3 in a rat model. PAH is the prototypical substrate of the renal organic anion transport system. Male Wistar rats underwent a bile duct ligation (BDL rats). Pair-fed sham-operated rats served as controls. BDL rats displayed a significantly lower systemic PAH clearance. Renal studies revealed a reduction in the renal clearance and in the excreted and secreted load of PAH in BDL rats. The OAT1 protein expression in kidney homogenates was not modified, but it decreased in the basolateral membranes from BDL rats. In contrast, OAT3 abundance in both kidney cortex homogenates and in basolateral membranes increased by 3 days after the ligation. Immunocytochemical studies (light microscopic and confocal immunofluorescence microscopic analyses) confirmed the changes in the renal expression of these transport proteins. The present study demonstrates the key role of OAT1 expression in the impaired elimination of PAH after 3 days of obstructive cholestasis.
Collapse
Affiliation(s)
- Anabel Brandoni
- Farmacología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, CONICET, Argentina
| | | | | | | | | |
Collapse
|
29
|
Miyazaki H, Anzai N, Ekaratanawong S, Sakata T, Shin HJ, Jutabha P, Hirata T, He X, Nonoguchi H, Tomita K, Kanai Y, Endou H. Modulation of Renal Apical Organic Anion Transporter 4 Function by Two PDZ Domain–Containing Proteins. J Am Soc Nephrol 2005; 16:3498-506. [PMID: 16236806 DOI: 10.1681/asn.2005030306] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Human organic anion transporter 4 (OAT4) is an apical organic anion/dicarboxylate exchanger in the renal proximal tubules and mediates high-affinity transport of steroid sulfates such as estrone-3-sulfate (E1S) and dehydroepiandrosterone sulfate. Here, two multivalent PDZ (PSD-95/Discs Large/ZO-1) proteins PDZK1 and NHERF1 were examined as interactors of OAT4 by a yeast two-hybrid assay. These interactions require the extreme C-terminal region of OAT4 and the first and fourth PDZ domains of PDZK1 and the first PDZ domain of NHERF1. These interactions were confirmed by surface plasmon resonance assays (K(D): 36 nM, 1.2 microM, and 41.7 microM, respectively). In vitro binding assays and co-immunoprecipitation studies revealed that the OAT4 wild-type but not a mutant lacking the PDZ motif interacted directly with both PDZK1 and NHERF1. OAT4, PDZK1, and NHERF1 proteins were shown to be localized at the apical membrane of renal proximal tubules. The association with PDZK1 or NHERF1 enhanced OAT4-mediated E1S transport activities in HEK293 cells (1.2- to 1.4-fold), and the deletion of the OAT4 C-terminal PDZ motif abolished this effect. The augmentation of the transport activity was accompanied by alteration in V(max) of E(1)S transport via OAT4 and was associated with the increased surface expression level of OAT4 protein. This study indicates that the functional activity of OAT4 is modulated through the PDZ interaction with the network of PDZK1 and NHERF1 and suggests that OAT4 is involved in the regulated apical organic anion handling in the renal proximal tubules, provided by the PDZ scaffold.
Collapse
Affiliation(s)
- Hiroki Miyazaki
- Department of Pharmacology and Toxicology, Kyorin University School of Medicine, Mitaka-shi, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|