1
|
Batarni S, Nayak N, Chang A, Li F, Hareendranath S, Zhou L, Xu H, Stroud R, Eriksen J, Edwards RH. Substrate recognition and proton coupling by a bacterial member of solute carrier family 17. J Biol Chem 2023; 299:104646. [PMID: 36965620 PMCID: PMC10149257 DOI: 10.1016/j.jbc.2023.104646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023] Open
Abstract
The solute carrier 17 family transports diverse organic anions using two distinct modes of coupling to a source of energy. Transporters that package glutamate and nucleotide into secretory vesicles for regulated release by exocytosis are driven by membrane potential but subject to allosteric regulation by H+ and Cl-. Other solute carrier 17 members including the lysosomal sialic acid exporter couple the flux of organic anion to cotransport of H+. To begin to understand how similar proteins can perform such different functions, we have studied Escherichia coli DgoT, a H+/galactonate cotransporter. A recent structure of DgoT showed many residues contacting D-galactonate, and we now find that they do not tolerate even conservative substitutions. In contrast, the closely related lysosomal H+/sialic acid cotransporter Sialin tolerates similar mutations, consistent with its recognition of diverse substrates with relatively low affinity. We also find that despite coupling to H+, DgoT transports more rapidly but with lower apparent affinity at high pH. Indeed, membrane potential can drive uptake, indicating electrogenic transport and suggesting a H+:galactonate stoichiometry >1. Located in a polar pocket of the N-terminal helical bundle, Asp46 and Glu133 are each required for net flux by DgoT, but the E133Q mutant exhibits robust exchange activity and rescues exchange by D46N, suggesting that these two residues operate in series to translocate protons. E133Q also shifts the pH sensitivity of exchange by DgoT, supporting a central role for the highly conserved TM4 glutamate in H+ coupling by DgoT.
Collapse
Affiliation(s)
- Samir Batarni
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, California
| | - Nanda Nayak
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, California
| | - Audrey Chang
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, California
| | - Fei Li
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, California
| | - Surabhi Hareendranath
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, California
| | - Lexi Zhou
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, California
| | - Hongfei Xu
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, California
| | - Robert Stroud
- Department of Biochemistry & Biophysics, UCSF School of Medicine, San Francisco, California
| | - Jacob Eriksen
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, California.
| | - Robert H Edwards
- Departments of Neurology and Physiology, UCSF School of Medicine, San Francisco, California.
| |
Collapse
|
2
|
How the diagnosis and the management of genetic renal phosphate leak impact the life of kidney stone formers? Urolithiasis 2022; 50:319-331. [DOI: 10.1007/s00240-022-01316-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 02/08/2022] [Indexed: 10/19/2022]
|
3
|
You Y, Ren Y, Liu J, Qu J. Promising Epigenetic Biomarkers Associated With Cancer-Associated-Fibroblasts for Progression of Kidney Renal Clear Cell Carcinoma. Front Genet 2021; 12:736156. [PMID: 34630525 PMCID: PMC8495159 DOI: 10.3389/fgene.2021.736156] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) is the most common malignant kidney tumor as its characterization of highly metastatic potential. Patients with KIRC are associated with poor clinical outcomes with limited treatment options. Up to date, the underlying molecular mechanisms of KIRC pathogenesis and progression are still poorly understood. Instead, particular features of Cancer-Associated Fibroblasts (CAFs) are highly associated with adverse outcomes of patients with KIRC, while the precise regulatory mechanisms at the epigenetic level of KIRC in governing CAFs remain poorly defined. Therefore, explore the correlations between epigenetic regulation and CAFs infiltration may help us better understand the molecular mechanisms behind KIRC progression, which may improve clinical outcomes and patients quality of life. In the present study, we identified a set of clinically relevant CAFs-related methylation-driven genes, NAT8, TINAG, and SLC17A1 in KIRC. Our comprehensive in silico analysis revealed that the expression levels of NAT8, TINAG, and SLC17A1 are highly associated with outcomes of patients with KIRC. Meanwhile, their methylation levels are highly correlates with the severity of KIRC. We suggest that the biomarkers might contribute to CAFs infiltration in KIRC. Taken together, our study provides a set of promising biomarkers which could predict the progression and prognosis of KIRC. Our findings could have potential prognosis and therapeutic significance in the progression of KIRC.
Collapse
Affiliation(s)
- Yongke You
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen, China
| | - Yeping Ren
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen, China
| | - Jikui Liu
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jianhua Qu
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
4
|
Eriksen J, Li F, Edwards RH. The mechanism and regulation of vesicular glutamate transport: Coordination with the synaptic vesicle cycle. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183259. [PMID: 32147354 DOI: 10.1016/j.bbamem.2020.183259] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 01/30/2023]
Abstract
The transport of classical neurotransmitters into synaptic vesicles generally relies on a H+ electrochemical gradient (∆μH+). Synaptic vesicle uptake of glutamate depends primarily on the electrical component ∆ψ as the driving force, rather than the chemical component ∆pH. However, the vesicular glutamate transporters (VGLUTs) belong to the solute carrier 17 (SLC17) family, which includes closely related members that function as H+ cotransporters. Recent work has also shown that the VGLUTs undergo allosteric regulation by H+ and Cl-, and exhibit an associated Cl- conductance. These properties appear to coordinate VGLUT activity with the large ionic shifts that accompany the rapid recycling of synaptic vesicles driven by neural activity. Recent structural information also suggests common mechanisms that underlie the apparently divergent function of SLC17 family members, and that confer allosteric regulation.
Collapse
Affiliation(s)
- Jacob Eriksen
- Department of Physiology, UCSF School of Medicine, United States of America; Department of Neurology, UCSF School of Medicine, United States of America
| | - Fei Li
- Department of Physiology, UCSF School of Medicine, United States of America; Department of Neurology, UCSF School of Medicine, United States of America
| | - Robert H Edwards
- Department of Physiology, UCSF School of Medicine, United States of America; Department of Neurology, UCSF School of Medicine, United States of America.
| |
Collapse
|
5
|
Wubuli A, Reyer H, Muráni E, Ponsuksili S, Wolf P, Oster M, Wimmers K. Tissue-Wide Gene Expression Analysis of Sodium/Phosphate Co-Transporters in Pigs. Int J Mol Sci 2019; 20:ijms20225576. [PMID: 31717287 PMCID: PMC6888643 DOI: 10.3390/ijms20225576] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/01/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022] Open
Abstract
Sodium/phosphate co-transporters are considered to be important mediators of phosphorus (P) homeostasis. The expression of specific sodium/phosphate co-transporters is routinely used as an immediate response to dietary interventions in different species. However, a general understanding of their tissue-specificity is required to elucidate their particular contribution to P homeostasis. In this study, the tissue-wide gene expression status of all currently annotated sodium/phosphate co-transporters were investigated in two pig trials focusing on a standard commercial diet (trial 1) or divergent P-containing diets (trial 2). A wide range of tissues including the gastrointestinal tract (stomach, duodenum, jejunum, ileum, caecum, and colon), kidney, liver, bone, muscle, lung, and aorta were analyzed. Both trials showed consistent patterns in the overall tissue-specific expression of P transporters. While SLC34A2 was considered as the most important intestinal P transporter in other species including humans, SLC34A3 appeared to be the most prominent intestinal P transporter in pigs. In addition, the P transporters of the SLC17 family showed basal expression in the pig intestine and might have a contribution to P homeostasis. The expression patterns observed in the distal colon provide evidence that the large intestine may also be relevant for intestinal P absorption. A low dietary P supply induced higher expressions of SLC20A1, SLC20A2, SLC34A1, and SLC34A3 in the kidney cortex. The results suggest that the expression of genes encoding transcellular P transporters is tissue-specific and responsive to dietary P supply, while underlying regulatory mechanisms require further analyses.
Collapse
Affiliation(s)
- Aisanjiang Wubuli
- Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.W.); (H.R.); (E.M.); (S.P.); (M.O.)
| | - Henry Reyer
- Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.W.); (H.R.); (E.M.); (S.P.); (M.O.)
| | - Eduard Muráni
- Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.W.); (H.R.); (E.M.); (S.P.); (M.O.)
| | - Siriluck Ponsuksili
- Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.W.); (H.R.); (E.M.); (S.P.); (M.O.)
| | - Petra Wolf
- Nutrition Physiology and Animal Nutrition, University of Rostock, Justus-von-Liebig-Weg 6b, 18059 Rostock, Germany;
| | - Michael Oster
- Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.W.); (H.R.); (E.M.); (S.P.); (M.O.)
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany; (A.W.); (H.R.); (E.M.); (S.P.); (M.O.)
- Animal Breeding and Genetics, University of Rostock, Justus-von-Liebig-Weg 7, 18059 Rostock, Germany
- Correspondence: ; Tel.: +49-38208-68600
| |
Collapse
|
6
|
Abstract
Phosphate is essential for skeletal mineralization, and its chronic deficiency leads to rickets and osteomalacia. Skeletal mineralization starts in matrix vesicles (MVs) derived from the plasma membrane of osteoblasts and chondrocytes. MVs contain high activity of tissue non-specific alkaline phosphatase (TNSALP), which hydrolyzes phosphoric esters such as pyrophosphates (PPi) to produce inorganic orthophosphates (Pi). Extracellular Pi in the skeleton is taken up by MVs through type III sodium/phosphate (Na+/Pi) cotransporters and forms hydroxyapatite. In addition to its roles in MV-mediated skeletal mineralization, accumulating evidence has revealed that extracellular Pi evokes signal transduction and regulates cellular function. Pi induces apoptosis of hypertrophic chondrocytes, which is a critical step for endochondral ossification. Extracellular Pi also regulates the expression of various genes including those related to proliferation, differentiation, and mineralization. In vitro cell studies have demonstrated that an elevation in extracellular Pi level leads to the activation of fibroblast growth factor receptor (FGFR), Raf/MEK (mitogen-activated protein kinase/ERK kinase)/ERK (extracellular signal-regulated kinase) pathway, where the type III Na+/Pi cotransporter PiT-1 may be involved. Responsiveness of skeletal cells to extracellular Pi suggests their ability to sense and adapt to an alteration in Pi availability in their environment. Involvement of FGFR in the Pi-evoked signal transduction is interesting because enhanced FGFR signaling in osteoblasts/osteocytes might be responsible for the overproduction of FGF23, a key molecule in phosphate homeostasis, in a mouse model for human X-linked hypophosphatemic rickets (XLH). Impaired Pi sensing may be a pathogenesis of XLH, which needs to be clarified in future.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, Izumi, Japan
- *Correspondence: Toshimi Michigami
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
7
|
Phosphate transport: from microperfusion to molecular cloning. Pflugers Arch 2018; 471:1-6. [PMID: 30569199 DOI: 10.1007/s00424-018-2245-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022]
|
8
|
Michigami T, Kawai M, Yamazaki M, Ozono K. Phosphate as a Signaling Molecule and Its Sensing Mechanism. Physiol Rev 2018; 98:2317-2348. [DOI: 10.1152/physrev.00022.2017] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In mammals, phosphate balance is maintained by influx and efflux via the intestines, kidneys, bone, and soft tissue, which involves multiple sodium/phosphate (Na+/Pi) cotransporters, as well as regulation by several hormones. Alterations in the levels of extracellular phosphate exert effects on both skeletal and extra-skeletal tissues, and accumulating evidence has suggested that phosphate itself evokes signal transduction to regulate gene expression and cell behavior. Several in vitro studies have demonstrated that an elevation in extracellular Piactivates fibroblast growth factor receptor, Raf/MEK (mitogen-activated protein kinase/ERK kinase)/ERK (extracellular signal-regulated kinase) pathway and Akt pathway, which might involve the type III Na+/Picotransporter PiT-1. Excessive phosphate loading can lead to various harmful effects by accelerating ectopic calcification, enhancing oxidative stress, and dysregulating signal transduction. The responsiveness of mammalian cells to altered extracellular phosphate levels suggests that they may sense and adapt to phosphate availability, although the precise mechanism for phosphate sensing in mammals remains unclear. Unicellular organisms, such as bacteria and yeast, use some types of Pitransporters and other molecules, such as kinases, to sense the environmental Piavailability. Multicellular animals may need to integrate signals from various organs to sense the phosphate levels as a whole organism, similarly to higher plants. Clarification of the phosphate-sensing mechanism in humans may lead to the development of new therapeutic strategies to prevent and treat diseases caused by phosphate imbalance.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masanobu Kawai
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Miwa Yamazaki
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Keiichi Ozono
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
9
|
Preobraschenski J, Cheret C, Ganzella M, Zander JF, Richter K, Schenck S, Jahn R, Ahnert-Hilger G. Dual and Direction-Selective Mechanisms of Phosphate Transport by the Vesicular Glutamate Transporter. Cell Rep 2018; 23:535-545. [DOI: 10.1016/j.celrep.2018.03.055] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 02/13/2018] [Accepted: 03/14/2018] [Indexed: 11/30/2022] Open
|
10
|
Vadakedath S, Kandi V. Probable Potential Role of Urate Transporter Genes in the Development of Metabolic Disorders. Cureus 2018; 10:e2382. [PMID: 29850377 PMCID: PMC5973493 DOI: 10.7759/cureus.2382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Metabolic disorders are a group of interrelated conditions which increases the risk of developing heart diseases, stroke, and diabetes. These usually occur as a consequence of deficiency of enzymes involved in biochemical reactions in the body. The dietary habits, lack of physical exercise, stress, and genetic susceptibility leads to an increased risk of developing metabolic disorders. A diet rich in processed food items containing high calories aggravates the production of a purine metabolite, the uric acid (UA). UA functions as an antioxidant, protects against inflammation, aging, and cancer. It exists as urate ions in the circulation and blood level of UA is maintained by a balance between its production in the liver and its excretion by the renal tubules. The regular excretion of UA through the kidneys is necessary to maintain optimum blood levels of UA (3-7 mg/dl). There are various transporters of uric acid present around the renal tubules, which help in reabsorption of UA into the blood. These urate transporters (UT) are proteins coded in the genes. Mutations in these genes may prompt disturbances in UA reabsorption, and could lead to the development of hyperuricaemia, insulin resistance, endothelial dysfunction, diabetes and other metabolic diseases. This paper reviews eight such genes coding for UTs and attempts to unravel the link between the activities of UA, UTs, and the consequences during mutations in the genes coding for the UTs in the development of metabolic disorders. The genes reviewed included SLC2A9, SLC17A1, SLC22A12, SLC16A9, GCKR, LRRC16A, PDZK1, and ABCG2.
Collapse
|
11
|
Eriksen J, Chang R, McGregor M, Silm K, Suzuki T, Edwards RH. Protons Regulate Vesicular Glutamate Transporters through an Allosteric Mechanism. Neuron 2016; 90:768-80. [PMID: 27133463 DOI: 10.1016/j.neuron.2016.03.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 02/25/2016] [Accepted: 03/21/2016] [Indexed: 11/25/2022]
Abstract
The quantal nature of synaptic transmission requires a mechanism to transport neurotransmitter into synaptic vesicles without promoting non-vesicular efflux across the plasma membrane. Indeed, the vesicular transport of most classical transmitters involves a mechanism of H(+) exchange, which restricts flux to acidic membranes such as synaptic vesicles. However, vesicular transport of the principal excitatory transmitter glutamate depends primarily on membrane potential, which would drive non-vesicular efflux, and the role of protons is unclear. Adapting electrophysiology to record currents associated with the vesicular glutamate transporters (VGLUTs), we characterize a chloride conductance that is gated by lumenal protons and chloride and supports glutamate uptake. Rather than coupling stoichiometrically to glutamate flux, lumenal protons and chloride allosterically activate vesicular glutamate transport. Gating by protons serves to inhibit what would otherwise be substantial non-vesicular glutamate efflux at the plasma membrane, thereby restricting VGLUT activity to synaptic vesicles.
Collapse
Affiliation(s)
- Jacob Eriksen
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Roger Chang
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA; Graduate Program in Biomedical Sciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Matt McGregor
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA; Graduate Program in Neuroscience, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Katlin Silm
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Toshiharu Suzuki
- Faculty of Science and Engineering, Waseda University, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Robert H Edwards
- Department of Physiology, UCSF School of Medicine, San Francisco, CA 94143, USA; Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143, USA; Graduate Program in Biomedical Sciences, UCSF School of Medicine, San Francisco, CA 94143, USA; Graduate Program in Neuroscience, UCSF School of Medicine, San Francisco, CA 94143, USA.
| |
Collapse
|
12
|
Vesicular glutamate transporters as anion channels? Pflugers Arch 2015; 468:513-8. [DOI: 10.1007/s00424-015-1760-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/05/2015] [Accepted: 11/12/2015] [Indexed: 01/13/2023]
|
13
|
Koyama T, Matsui D, Kuriyama N, Ozaki E, Tanaka K, Oze I, Hamajima N, Wakai K, Okada R, Arisawa K, Mikami H, Shimatani K, Hirata A, Takashima N, Suzuki S, Nagata C, Kubo M, Tanaka H. Genetic variants of SLC17A1 are associated with cholesterol homeostasis and hyperhomocysteinaemia in Japanese men. Sci Rep 2015; 5:15888. [PMID: 26524967 PMCID: PMC4630628 DOI: 10.1038/srep15888] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
Abstract
Hyperuricaemia is an undisputed and highly predictive biomarker for cardiovascular risk. SLC17A1, expressed in the liver and kidneys, harbours potent candidate single nucleotide polymorphisms that decrease uric acid levels. Therefore, we examined SLC17A1 polymorphisms (rs1165196, rs1179086, and rs3757131), which might suppress cardiovascular risk factors and that are involved in liver functioning, via a large-scale pooled analysis of the Japanese general population in a cross-sectional study. Using data from the Japan Multi-Institutional Collaborative Cohort Study, we identified 1842 participants of both sexes, 35–69-years-old, having the requisite data, and analysed their SLC17A1 genotypes. In men, logistic regression analyses revealed that minor alleles in SLC17A1 polymorphisms (rs1165196 and rs3757131) were associated with a low-/high-density lipoprotein cholesterol ratio >2.0 (rs1165196: odds ratio [OR], 0.703; 95% confidence interval [CI], 0.536–0.922; rs3757131: OR, 0.658; 95% CI, 0.500–0.866), and with homocysteine levels of >10.0 nmol/mL (rs1165196: OR, 0.544; 95% CI, 0.374–0.792; rs3757131: OR, 0.509; 95% CI, 0.347–0.746). Therefore, these polymorphisms had dominant negative effects on cholesterol homeostasis and hyperhomocysteinaemia, in men, independent of alcohol consumption, physical activity, or daily energy and nutrition intake. Thus, genetic variants of SLC17A1 are potential biomarkers for altered cholesterol homeostasis and hyperhomocysteinaemia in Japanese men.
Collapse
Affiliation(s)
- Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Matsui
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nagato Kuriyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Etsuko Ozaki
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keitaro Tanaka
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Isao Oze
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Nobuyuki Hamajima
- Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kokichi Arisawa
- Department of Preventive Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Haruo Mikami
- Division of Cancer Registry, Prevention and Epidemiology, Chiba Cancer Center, Chiba, Japan
| | - Keiichi Shimatani
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Akie Hirata
- Department of Geriatric Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoyuki Takashima
- Department of Public Health, Shiga University of Medical Science, Otsu, Japan
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Chisato Nagata
- Department of Epidemiology and Preventive Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Michiaki Kubo
- Laboratory for Genotyping Development, Center for Genomic Medicine, RIKEN, Yokohama, Japan
| | - Hideo Tanaka
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| |
Collapse
|
14
|
Omote H, Miyaji T, Hiasa M, Juge N, Moriyama Y. Structure, Function, and Drug Interactions of Neurotransmitter Transporters in the Postgenomic Era. Annu Rev Pharmacol Toxicol 2015; 56:385-402. [PMID: 26514205 DOI: 10.1146/annurev-pharmtox-010814-124816] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Vesicular neurotransmitter transporters are responsible for the accumulation of neurotransmitters in secretory vesicles and play essential roles in chemical transmission. The SLC17 family contributes to sequestration of anionic neurotransmitters such as glutamate, aspartate, and nucleotides. Identification and subsequent cellular and molecular biological studies of SLC17 transporters unveiled the principles underlying the actions of these transporters. Recent progress in reconstitution methods in combination with postgenomic approaches has advanced studies on neurotransmitter transporters. This review summarizes the molecular properties of SLC17-type transporters and recent findings regarding the novel SLC18 transporter.
Collapse
Affiliation(s)
- Hiroshi Omote
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan; ,
| | - Takaaki Miyaji
- Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| | - Miki Hiasa
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan; ,
| | - Narinobu Juge
- Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| | - Yoshinori Moriyama
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan; , .,Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
15
|
Pujol-Giménez J, Pérez A, Reyes AM, Loo DDF, Lostao MP. Functional characterization of the human facilitative glucose transporter 12 (GLUT12) by electrophysiological methods. Am J Physiol Cell Physiol 2015; 308:C1008-22. [PMID: 25855082 DOI: 10.1152/ajpcell.00343.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 04/06/2015] [Indexed: 12/11/2022]
Abstract
GLUT12 is a member of the facilitative family of glucose transporters. The goal of this study was to characterize the functional properties of GLUT12, expressed in Xenopus laevis oocytes, using radiotracer and electrophysiological methods. Our results showed that GLUT12 is a facilitative sugar transporter with substrate selectivity: d-glucose ≥ α-methyl-d-glucopyranoside (α-MG) > 2-deoxy-d-glucose(2-DOG) > d-fructose = d-galactose. α-MG is a characteristic substrate of the Na(+)/glucose (SGLT) family and has not been shown to be a substrate of any of the GLUTs. In the absence of sugar, (22)Na(+) was transported through GLUT12 at a higher rate (40%) than noninjected oocytes, indicating that there is a Na(+) leak through GLUT12. Genistein, an inhibitor of GLUT1, also inhibited sugar uptake by GLUT12. Glucose uptake was increased by the PKA activator 8-bromoadenosine 3',5'-cyclic monophosphate (8-Br-cAMP) but not by the PKC activator phorbol-12-myristate-13-acetate (PMA). In high K(+) concentrations, glucose uptake was blocked. Addition of glucose to the external solution induced an inward current with a reversal potential of approximately -15 mV and was blocked by Cl(-) channel blockers, indicating the current was carried by Cl(-) ions. The sugar-activated Cl(-) currents were unaffected by genistein. In high external K(+) concentrations, sugar-activated Cl(-) currents were also blocked, indicating that GLUT12 activity is voltage dependent. Furthermore, glucose-induced current was increased by the PKA activator 8-Br-cAMP but not by the PKC activator PMA. These new features of GLUT12 are very different from those described for other GLUTs, indicating that GLUT12 must have a specific physiological role within glucose homeostasis, still to be discovered.
Collapse
Affiliation(s)
- Jonai Pujol-Giménez
- Department of Nutrition, Food Science and Physiology, School of Pharmacy, University of Navarra, Pamplona, Spain
| | - Alejandra Pérez
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile; and
| | - Alejandro M Reyes
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile; and
| | - Donald D F Loo
- Department of Physiology, David Geffen School of Medicine at University of California, Los Angeles, California
| | - Maria Pilar Lostao
- Department of Nutrition, Food Science and Physiology, School of Pharmacy, University of Navarra, Pamplona, Spain;
| |
Collapse
|
16
|
Biber J, Murer H, Mohebbi N, Wagner C. Renal Handling of Phosphate and Sulfate. Compr Physiol 2014; 4:771-92. [DOI: 10.1002/cphy.c120031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: transporters. Br J Pharmacol 2013; 170:1706-96. [PMID: 24528242 PMCID: PMC3892292 DOI: 10.1111/bph.12450] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Transporters are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
18
|
Haller M, Amatschek S, Wilflingseder J, Kainz A, Bielesz B, Pavik I, Serra A, Mohebbi N, Biber J, Wagner CA, Oberbauer R. Sirolimus induced phosphaturia is not caused by inhibition of renal apical sodium phosphate cotransporters. PLoS One 2012; 7:e39229. [PMID: 22859939 PMCID: PMC3408497 DOI: 10.1371/journal.pone.0039229] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 05/17/2012] [Indexed: 12/01/2022] Open
Abstract
The vast majority of glomerular filtrated phosphate is reabsorbed in the proximal tubule. Posttransplant phosphaturia is common and aggravated by sirolimus immunosuppression. The cause of sirolimus induced phosphaturia however remains elusive. Male Wistar rats received sirolimus or vehicle for 2 or 7 days (1.5mg/kg). The urine phosphate/creatinine ratio was higher and serum phosphate was lower in sirolimus treated rats, fractional excretion of phosphate was elevated and renal tubular phosphate reabsorption was reduced suggesting a renal cause for hypophosphatemia. PTH was lower in sirolimus treated rats. FGF 23 levels were unchanged at day 2 but lower in sirolimus treated rats after 7 days. Brush border membrane vesicle phosphate uptake was not altered in sirolimus treated groups or by direct incubation with sirolimus. mRNA, protein abundance, and subcellular transporter distribution of NaPi-IIa, Pit-2 and NHE3 were not different between groups but NaPi-IIc mRNA expression was lower at day 7. Transcriptome analyses revealed candidate genes that could be involved in the phosphaturic response. Sirolimus caused a selective renal phosphate leakage, which was not mediated by NaPi-IIa or NaPi-IIc regulation or localization. We hypothesize that another mechanism such as a basolateral phosphate transporter may be responsible for the sirolimus induced phosphaturia.
Collapse
Affiliation(s)
- Maria Haller
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
- Institute of Physiology, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- Department of Nephrology and Transplantation, KH Elisabethinen Linz, Linz, Austria
| | - Stefan Amatschek
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
| | | | - Alexander Kainz
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
| | - Bernd Bielesz
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
| | - Ivana Pavik
- Institute of Physiology, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Andreas Serra
- Institute of Physiology, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Nilufar Mohebbi
- Institute of Physiology, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Jürg Biber
- Institute of Physiology, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Carsten A. Wagner
- Institute of Physiology, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Rainer Oberbauer
- Department of Nephrology, Medical University of Vienna, Vienna, Austria
- Department of Nephrology and Transplantation, KH Elisabethinen Linz, Linz, Austria
- Austrian Dialysis and Transplant Registry, Linz, Austria
- * E-mail:
| |
Collapse
|
19
|
Hollis-Moffatt JE, Phipps-Green AJ, Chapman B, Jones GT, van Rij A, Gow PJ, Harrison AA, Highton J, Jones PB, Montgomery GW, Stamp LK, Dalbeth N, Merriman TR. The renal urate transporter SLC17A1 locus: confirmation of association with gout. Arthritis Res Ther 2012; 14:R92. [PMID: 22541845 PMCID: PMC3446466 DOI: 10.1186/ar3816] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 04/09/2012] [Accepted: 04/27/2012] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Two major gout-causing genes have been identified, the urate transport genes SLC2A9 and ABCG2. Variation within the SLC17A1 locus, which encodes sodium-dependent phosphate transporter 1, a renal transporter of uric acid, has also been associated with serum urate concentration. However, evidence for association with gout is equivocal. We investigated the association of the SLC17A1 locus with gout in New Zealand sample sets. METHODS Five variants (rs1165196, rs1183201, rs9358890, rs3799344, rs12664474) were genotyped across a New Zealand sample set totaling 971 cases and 1,742 controls. Cases were ascertained according to American Rheumatism Association criteria. Two population groups were studied: Caucasian and Polynesian. RESULTS At rs1183201 (SLC17A1), evidence for association with gout was observed in both the Caucasian (odds ratio (OR) = 0.67, P = 3.0 × 10-6) and Polynesian (OR = 0.74, P = 3.0 × 10-3) groups. Meta-analysis confirmed association of rs1183201 with gout at a genome-wide level of significance (OR = 0.70, P = 3.0 × 10-8). Haplotype analysis suggested the presence of a common protective haplotype. CONCLUSION We confirm the SLC17A1 locus as the third associated with gout at a genome-wide level of significance.
Collapse
Affiliation(s)
- Jade E Hollis-Moffatt
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin 9054, New Zealand
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lederer E, Miyamoto KI. Clinical consequences of mutations in sodium phosphate cotransporters. Clin J Am Soc Nephrol 2012; 7:1179-87. [PMID: 22516291 DOI: 10.2215/cjn.09090911] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Three families of sodium phosphate cotransporters have been described. Their specific roles in human health and disease have not been defined. Review of the literature reveals that the type II sodium phosphate cotransporters play a significant role in transepithelial transport in a number of tissues including kidney, intestine, salivary gland, mammary gland, and lung. The type I transporters seem to play a major role in renal urate handling and mutations in these proteins have been implicated in susceptibility to gout. The ubiquitously expressed type III transporters play a lesser role in phosphate homeostasis but contribute to cellular phosphate uptake, mineralization, and inflammation. The recognition of species differences in the expression, regulation, and function of these transport proteins suggests an urgent need to find ways to study them in humans.
Collapse
Affiliation(s)
- Eleanor Lederer
- University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | | |
Collapse
|
21
|
Pietrancosta N, Anne C, Prescher H, Ruivo R, Sagné C, Debacker C, Bertrand HO, Brossmer R, Acher F, Gasnier B. Successful prediction of substrate-binding pocket in SLC17 transporter sialin. J Biol Chem 2012; 287:11489-97. [PMID: 22334707 DOI: 10.1074/jbc.m111.313056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Secondary active transporters from the SLC17 protein family are required for excitatory and purinergic synaptic transmission, sialic acid metabolism, and renal function, and several members are associated with inherited neurological or metabolic diseases. However, molecular tools to investigate their function or correct their genetic defects are limited or absent. Using structure-activity, homology modeling, molecular docking, and mutagenesis studies, we have located the substrate-binding site of sialin (SLC17A5), a lysosomal sialic acid exporter also recently implicated in exocytotic release of aspartate. Human sialin is defective in two inherited sialic acid storage diseases and is responsible for metabolic incorporation of the dietary nonhuman sialic acid N-glycolylneuraminic acid. We built cytosol-open and lumen-open three-dimensional models of sialin based on weak, but significant, sequence similarity with the glycerol-3-phosphate and fucose permeases from Escherichia coli, respectively. Molecular docking of 31 synthetic sialic acid analogues to both models was consistent with inhibition studies. Narrowing the sialic acid-binding site in the cytosol-open state by two phenylalanine to tyrosine mutations abrogated recognition of the most active analogue without impairing neuraminic acid transport. Moreover, a pilot virtual high-throughput screening of the cytosol-open model could identify a pseudopeptide competitive inhibitor showing >100-fold higher affinity than the natural substrate. This validated model of human sialin and sialin-guided models of other SLC17 transporters should pave the way for the identification of inhibitors, glycoengineering tools, pharmacological chaperones, and fluorescent false neurotransmitters targeted to these proteins.
Collapse
Affiliation(s)
- Nicolas Pietrancosta
- Centre National de la Recherche Scientifique, UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, F-75006 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Blakely RD, Edwards RH. Vesicular and plasma membrane transporters for neurotransmitters. Cold Spring Harb Perspect Biol 2012; 4:a005595. [PMID: 22199021 PMCID: PMC3281572 DOI: 10.1101/cshperspect.a005595] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The regulated exocytosis that mediates chemical signaling at synapses requires mechanisms to coordinate the immediate response to stimulation with the recycling needed to sustain release. Two general classes of transporter contribute to release, one located on synaptic vesicles that loads them with transmitter, and a second at the plasma membrane that both terminates signaling and serves to recycle transmitter for subsequent rounds of release. Originally identified as the target of psychoactive drugs, these transport systems have important roles in transmitter release, but we are only beginning to understand their contribution to synaptic transmission, plasticity, behavior, and disease. Recent work has started to provide a structural basis for their activity, to characterize their trafficking and potential for regulation. The results indicate that far from the passive target of psychoactive drugs, neurotransmitter transporters undergo regulation that contributes to synaptic plasticity.
Collapse
Affiliation(s)
- Randy D Blakely
- Department of Pharmacology and Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548, USA
| | | |
Collapse
|
23
|
Forster IC, Hernando N, Biber J, Murer H. Phosphate transport kinetics and structure-function relationships of SLC34 and SLC20 proteins. CURRENT TOPICS IN MEMBRANES 2012. [PMID: 23177991 DOI: 10.1016/b978-0-12-394316-3.00010-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transport of inorganic phosphate (P(i)) is mediated by proteins belonging to two solute carrier families (SLC20 and SLC34). Members of both families transport P(i) using the electrochemical gradient for Na(+). The role of the SLC34 members as essential players in mammalian P(i) homeostasis is well established, whereas that of SLC20 proteins is less well defined. The SLC34 family comprises the following three isoforms that preferentially cotransport divalent P(i) and are expressed in epithelial tissue: the renal NaPi-IIa and NaPi-IIc are responsible for reabsorbing P(i) in the proximal tubule, whereas NaPi-IIb is more ubiquitously expressed, including the small intestine, where it mediates dietary P(i) absorption. The SLC20 family comprises two members (PiT-1, PiT-2) that preferentially cotransport monovalent P(i) and are expressed in epithelial as well as nonepithelial tissue. The transport kinetics of members of both families have been characterized in detail using heterologous expression in Xenopus oocytes. For the electrogenic NaPi-IIa/b, and PiT-1,-2, conventional electrophysiological techniques together with radiotracer methods have been applied, as well as time-resolved fluorometric measurements that allow new insights into local conformational changes of the protein during the cotransport cycle. For the electroneutral NaPi-IIc, conventional tracer uptake and fluorometry have been used to elucidate its transport properties. The 3-D structures of these proteins remain unresolved and structure-function studies have so far concentrated on defining the topology and identifying sites of functional importance.
Collapse
Affiliation(s)
- Ian C Forster
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Winterthurerstrasse, Zurich, Switzerland.
| | | | | | | |
Collapse
|
24
|
Abstract
Neurotransmitter identity is a defining feature of all neurons because it constrains the type of information they convey, but many neurons release multiple transmitters. Although the physiological role for corelease has remained poorly understood, the vesicular uptake of one transmitter can regulate filling with the other by influencing expression of the H(+) electrochemical driving force. In addition, the sorting of vesicular neurotransmitter transporters and other synaptic vesicle proteins into different vesicle pools suggests the potential for distinct modes of release. Corelease thus serves multiple roles in synaptic transmission.
Collapse
Affiliation(s)
- Thomas S Hnasko
- Departments of Physiology & Neurology, University of California, San Francisco, California 94158-2517, USA.
| | | |
Collapse
|
25
|
Omote H, Miyaji T, Juge N, Moriyama Y. Vesicular Neurotransmitter Transporter: Bioenergetics and Regulation of Glutamate Transport. Biochemistry 2011; 50:5558-65. [DOI: 10.1021/bi200567k] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Hiroshi Omote
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan
| | - Takaaki Miyaji
- Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| | - Narinobu Juge
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan
| | - Yoshinori Moriyama
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan
- Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
26
|
Miyamoto KI, Haito-Sugino S, Kuwahara S, Ohi A, Nomura K, Ito M, Kuwahata M, Kido S, Tatsumi S, Kaneko I, Segawa H. Sodium-dependent phosphate cotransporters: lessons from gene knockout and mutation studies. J Pharm Sci 2011; 100:3719-30. [PMID: 21567407 DOI: 10.1002/jps.22614] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 04/13/2011] [Accepted: 04/20/2011] [Indexed: 12/22/2022]
Abstract
Inorganic phosphate (Pi) is an essential physiological compound, highlighted by the syndromes caused by hypo or hyperphosphatemic states. Hyperphosphatemia is associated with ectopic calcification, cardiovascular disease, and increased mortality in patients with chronic kidney disease (CKD). As phosphate control is not efficient with diet or dialysis, oral Pi binders are used in over 90% of patients with renal failure. However, achieving tight control of serum Pi is difficult, and lower levels of serum Pi (severe hypophosphatemia) do not lead to better outcomes. The inhibition of sodium-dependent Pi (NaPi) transporter would be a preferable method to control serum Pi levels in patients with CKD or patients undergoing dialysis. Three types of NaPi transporters (types I-III) have been identified: solute carrier series SLC17A1 (NPT1/NaPi-I/OATv1), SLC34 (NaPi-IIa, NaPi-IIb, NaPi-IIc), and SLC20 (PiT1, PiT2), respectively. Knockout mice have been created for types I-III NaPi transporters. In this review, we discuss the roles of the NaPi transporters in Pi homeostasis.
Collapse
Affiliation(s)
- Ken-ichi Miyamoto
- Department of Molecular Nutrition, Institute of Health Biosciences, University of Tokushima Graduate School Tokushima 770-8503, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Courbebaisse M, Souberbielle JC. Équilibre phosphocalcique : régulation et explorations. Nephrol Ther 2011; 7:118-38. [DOI: 10.1016/j.nephro.2010.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
28
|
Yamazaki M, Ozono K, Okada T, Tachikawa K, Kondou H, Ohata Y, Michigami T. Both FGF23 and extracellular phosphate activate Raf/MEK/ERK pathway via FGF receptors in HEK293 cells. J Cell Biochem 2011; 111:1210-21. [PMID: 20717920 DOI: 10.1002/jcb.22842] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fibroblast growth factor 23 (FGF23) is a phosphaturic hormone produced by bone and exerts its function in the target organs by binding the FGF receptor (FGFR) and Klotho. Since recent studies suggested that extracellular inorganic phosphate (Pi) itself triggers signal transduction and regulates gene expression in some cell types, we tested the notion that extracellular Pi induces signal transduction in the target cells of FGF23 also and influences its signaling, utilizing a human embryonic kidney cell line HEK293. HEK293 cells expressed low levels of klotho, and treatment with a recombinant FGF23[R179Q], a proteolysis-resistant mutant of FGF23, resulted in phosphorylation of ERK1/2 and induction of early growth response-1 (EGR1) expression. Interestingly, increased extracellular Pi resulted in activation of the Raf/MEK/ERK pathway and expression of EGR1, which involved type III sodium/phosphate (Na(+)/Pi) cotransporter PiT-1. Since the effects of an inhibitor of Na(+)/Pi cotransporter on FGF23 signaling suggested that the signaling triggered by increased extracellular Pi shares the same downstream cascade as FGF23 signaling, we further investigated their convergence point. Increasing the extracellular Pi concentration resulted in the phosphorylation of FGF receptor substrate 2α (FRS2α), as did treatment with FGF23. Knockdown of FGFR1 expression diminished the phosphorylation of both FRS2α and ERK1/2 induced by the Pi. Moreover, overexpression of FGFR1 rescued the decrease in Pi-induced phosphorylation of ERK1/2 in the cells where the expression of PiT-1 was knocked down. These results suggest that increased extracellular Pi triggers signal transduction via PiT-1 and FGFR and influences FGF23 signaling in HEK293 cells.
Collapse
Affiliation(s)
- Miwa Yamazaki
- Department of Bone and Mineral Research, Osaka Medical Center and Research Institute for Maternal and Child Health, Izumi, Osaka 594-1101, Japan
| | | | | | | | | | | | | |
Collapse
|
29
|
Khoshniat S, Bourgine A, Julien M, Weiss P, Guicheux J, Beck L. The emergence of phosphate as a specific signaling molecule in bone and other cell types in mammals. Cell Mol Life Sci 2011; 68:205-18. [PMID: 20848155 PMCID: PMC11114507 DOI: 10.1007/s00018-010-0527-z] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 08/02/2010] [Accepted: 08/31/2010] [Indexed: 02/07/2023]
Abstract
Although considerable advances in our understanding of the mechanisms of phosphate homeostasis and skeleton mineralization have recently been made, little is known about the initial events involving the detection of changes in the phosphate serum concentrations and the subsequent downstream regulation cascade. Recent data has strengthened a long-established hypothesis that a phosphate-sensing mechanism may be present in various organs. Such a phosphate sensor would detect changes in serum or local phosphate concentration and would inform the body, the local environment, or the individual cell. This suggests that phosphate in itself could represent a signal regulating multiple factors necessary for diverse biological processes such as bone or vascular calcification. This review summarizes findings supporting the possibility that phosphate represents a signaling molecule, particularly in bone and cartilage, but also in other tissues. The involvement of various signaling pathways (ERK1/2), transcription factors (Fra-1, Runx2) and phosphate transporters (PiT1, PiT2) is discussed.
Collapse
Affiliation(s)
- Solmaz Khoshniat
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Annabelle Bourgine
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Marion Julien
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Pierre Weiss
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Jérôme Guicheux
- Group STEP (Skeletal Tissue Engineering and Physiopathology), Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), INSERM, U791, 44042 Nantes, France
- UFR Odontologie, Pres UNAM, 44042 Nantes, France
| | - Laurent Beck
- Growth and Signalling Research Center, INSERM, U845, 75015 Paris, France
- Faculté de Médecine, Centre de Recherche, INSERM U845, Université Paris Descartes, 156 Rue de Vaugirard, 75015 Paris, France
| |
Collapse
|
30
|
Abstract
Inherited diseases of renal phosphate handling lead to urinary phosphate wasting and depletion of total body phosphorus stores. Clinical sequelae of inherited disorders that are associated with increased urinary phosphate excretion are deleterious and can lead to abnormal skeletal growth and deformities. This Review describes hereditary disorders of renal phosphate wasting taking into account developments in our understanding of renal phosphate handling from the last decade. The cloning of genes involved in these disorders and further studies on their pathophysiological mechanisms have given important insights in to how phosphatonins, such as FGF-23, regulate renal phosphate reabsorption in health and disease. X-linked dominant hypophosphatemic rickets results from mutation of a metalloprotease (PHEX) that has an unidentified role in FGF-23 degradation. Mutation of an RXXR proteolytic cleavage site in FGF-23 prevents degradation and increases circulating levels of FGF-23 in autosomal dominant hypophosphatemic rickets. FGF-23 acts to remove sodium phosphate co-transporters from the luminal membrane of proximal tubular cells with resultant renal phosphate wasting. Loss of function mutations in genes encoding the transporters NaPi-IIc and NaPi-IIa also result in renal phosphate wasting and rickets.
Collapse
|
31
|
Regulation of renal sodium-dependent phosphate co-transporter genes (Npt2a and Npt2c) by all-trans-retinoic acid and its receptors. Biochem J 2010; 429:583-92. [DOI: 10.1042/bj20100484] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The type II sodium-dependent phosphate co-transporters Npt2a and Npt2c play critical roles in the reabsorption of Pi by renal proximal tubular cells. The vitamin A metabolite ATRA (all-trans-retinoic acid) is important for development, cell proliferation and differentiation, and bone formation. It has been reported that ATRA increases the rate of Pi transport in renal proximal tubular cells. However, the molecular mechanism is still unknown. In the present study, we observed the effects of a VAD (vitamin A-deficient) diet on Pi homoeostasis and the expression of Npt2a and Npt2c genes in rat kidney. There was no change in the plasma levels of Pi, but VAD rats significantly increased renal Pi excretion. Renal brush-border membrane Pi uptake activity and renal Npt2a and Npt2c expressions were significantly decreased in VAD rats. The transcriptional activity of a luciferase reporter plasmid containing the promoter region of human Npt2a and Npt2c genes was increased markedly by ATRA and a RAR (retinoic acid receptor)-specific analogue TTNPB {4-[E-2-(5,6,7,8-tetrahydro-5,5,8,8-tetra-methyl-2-naphtalenyl)-1-propenyl] benzoic acid} in renal proximal tubular cells overexpressing RARs and RXRs (retinoid X receptors). Furthermore, we identified RAREs (retinoic acid-response elements) in both gene promoters. Interestingly, the half-site sequences (5′-GGTTCA-3′: −563 to −558) of 2c-RARE1 overlapped the vitamin D-responsive element in the human Npt2c gene and were functionally important motifs for transcriptional regulation of human Npt2c by ATRA and 1,25(OH)2D3 (1α,25-dihydroxyvitamin D3), in both independent or additive actions. In summary, we conclude that VAD induces hyperphosphaturia through the down-regulation of Npt2a and Npt2c gene expression in the kidney.
Collapse
|
32
|
Affiliation(s)
- Dominique Prié
- Growth and Signaling Research Center and the Department of Physiology, INSERM Unité 845, Paris Descartes University, Paris, France
| | | |
Collapse
|
33
|
Iharada M, Miyaji T, Fujimoto T, Hiasa M, Anzai N, Omote H, Moriyama Y. Type 1 sodium-dependent phosphate transporter (SLC17A1 Protein) is a Cl(-)-dependent urate exporter. J Biol Chem 2010; 285:26107-13. [PMID: 20566650 DOI: 10.1074/jbc.m110.122721] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
SLC17A1 protein (NPT1) is the first identified member of the SLC17 phosphate transporter family and mediates the transmembrane cotransport of Na(+)/P(i) in oocytes. Although this protein is believed to be a renal polyspecific anion exporter, its transport properties are not well characterized. Here, we show that proteoliposomes containing purified SLC17A1 transport various organic anions such as p-aminohippuric acid and acetylsalicylic acid (aspirin) in an inside positive membrane potential (Deltapsi)-dependent manner. We found that NPT1 also transported urate. The uptake characteristics were similar to that of SLC17 members in its Cl(-) dependence and inhibitor sensitivity. When arginine 138, an essential amino acid residue for members of the SLC17 family such as the vesicular glutamate transporter, was specifically mutated to alanine, the resulting mutant protein was inactive in Deltapsi-dependent anion transport. Heterologously expressed and purified human NPT1 carrying the single nucleotide polymorphism mutation that is associated with increased risk of gout in humans exhibited 32% lower urate transport activity compared with the wild type protein. These results strongly suggested that NPT1 is a Cl(-)-dependent polyspecific anion exporter involved in urate excretion under physiological conditions.
Collapse
Affiliation(s)
- Masafumi Iharada
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Hnasko TS, Chuhma N, Zhang H, Goh GY, Sulzer D, Palmiter RD, Rayport S, Edwards RH. Vesicular glutamate transport promotes dopamine storage and glutamate corelease in vivo. Neuron 2010; 65:643-56. [PMID: 20223200 DOI: 10.1016/j.neuron.2010.02.012] [Citation(s) in RCA: 316] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2010] [Indexed: 01/03/2023]
Abstract
Dopamine neurons in the ventral tegmental area (VTA) play an important role in the motivational systems underlying drug addiction, and recent work has suggested that they also release the excitatory neurotransmitter glutamate. To assess a physiological role for glutamate corelease, we disrupted the expression of vesicular glutamate transporter 2 selectively in dopamine neurons. The conditional knockout abolishes glutamate release from midbrain dopamine neurons in culture and severely reduces their excitatory synaptic output in mesoaccumbens slices. Baseline motor behavior is not affected, but stimulation of locomotor activity by cocaine is impaired, apparently through a selective reduction of dopamine stores in the projection of VTA neurons to ventral striatum. Glutamate co-entry promotes monoamine storage by increasing the pH gradient that drives vesicular monoamine transport. Remarkably, low concentrations of glutamate acidify synaptic vesicles more slowly but to a greater extent than equimolar Cl(-), indicating a distinct, presynaptic mechanism to regulate quantal size.
Collapse
Affiliation(s)
- Thomas S Hnasko
- Departments of Physiology and Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Zuo L, Chen YR, Reyes LA, Lee HL, Chen CL, Villamena FA, Zweier JL. The radical trap 5,5-dimethyl-1-pyrroline N-oxide exerts dose-dependent protection against myocardial ischemia-reperfusion injury through preservation of mitochondrial electron transport. J Pharmacol Exp Ther 2009; 329:515-23. [PMID: 19201989 PMCID: PMC2672876 DOI: 10.1124/jpet.108.143479] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Accepted: 02/05/2009] [Indexed: 01/05/2023] Open
Abstract
Free radicals are important mediators of myocardial ischemia-reperfusion injury. Nitrone spin traps have been shown to scavenge free radicals. The cardioprotective effect of the spin trap, 5,5-dimethyl-1-pyrroline N-oxide (DMPO), was investigated in an isolated heart model of global ischemia and reperfusion. Rat hearts were perfused and subjected to global ischemia for 30 min followed by reperfusion with four treatment groups of varying DMPO concentration (0.5-10 mM) administered before induction of ischemia. DMPO treatment improved the recovery of left ventricular (LV) function and coronary flow over the 30-min period of reperfusion compared with untreated hearts. Enhanced recovery was observed for all doses studied but was highest with 1 mM treatment with 2.4-fold higher recovery of LV developed pressure and 37% reduction in infarct size. Superoxide was measured by tissue fluorometry using the O(2)* probe hydroethidine. Hearts treated with 1 mM DMPO showed a significant reduction in O(2)* production compared with control hearts both over the first 5 min of ischemia and upon reperfusion after 30 min of global ischemia. Studies of mitochondrial function demonstrated that 1 mM DMPO increased the recovery of function of complexes I, II/III, and IV after 30 min of reperfusion. Immunoblotting with antibodies against complexes I, II, and IV further revealed marked up-regulation of mitochondrial proteins, suggesting that DMPO prevents their ischemic degradation via scavenging oxygen radicals generated during ischemia/reperfusion. Thus, DMPO functions as a protective agent against ischemic and postischemic injury via radical scavenging, conferring robust dose-dependent protection with salvage of mitochondrial function and redox homeostasis.
Collapse
Affiliation(s)
- Li Zuo
- Center for Biomedical Electron Paramagnetic Resonance Spectroscopy and Imaging, The Davis Heart and Lung Research Institute, Ohio State University, Columbus, OH 43210-1252, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Cubero B, Nakagawa Y, Jiang XY, Miura KJ, Li F, Raghothama KG, Bressan RA, Hasegawa PM, Pardo JM. The phosphate transporter PHT4;6 is a determinant of salt tolerance that is localized to the Golgi apparatus of Arabidopsis. MOLECULAR PLANT 2009; 2:535-52. [PMID: 19825636 DOI: 10.1093/mp/ssp013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Insertion mutations that disrupt the function of PHT4;6 (At5g44370) cause NaCl hypersensitivity of Arabidopsis seedlings that is characterized by reduced growth of the primary root, enhanced lateral branching, and swelling of root tips. Mutant phenotypes were exacerbated by sucrose, but not by equiosmolar concentrations of mannitol, and attenuated by low inorganic phosphate in the medium. Protein PHT4;6 belongs to the Major Facilitator Superfamily of permeases that shares significant sequence similarity to mammalian type-I Pi transporters and vesicular glutamate transporters, and is a member of the PHT4 family of putative intracellular phosphate transporters of plants. PHT4;6 localizes to the Golgi membrane and transport studies indicate that PHT4;6 facilitates the selective transport of Pi but not of chloride or inorganic anions. Phenotypic similarities with other mutants displaying root swelling suggest that PHT4;6 likely functions in protein N-glycosylation and cell wall biosynthesis, which are essential for salt tolerance. Together, our results indicate that PHT4;6 transports Pi out of the Golgi lumenal space for the re-cycling of the Pi released from glycosylation processes.
Collapse
Affiliation(s)
- Beatriz Cubero
- Instituto de Recursos Naturales y Agrobiologia, Consejo Superior de Investigaciones Cientificas, Avda Reina Mercedes 10, Sevilla-41012, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
The kidney is a key player in phosphate balance. Inappropriate renal phosphate transport may alter serum phosphate concentration and bone mineralization, and increase the risk of renal lithiasis or soft tissue calcifications. The recent identification of fibroblast growth factor 23 (FGF23) as a hormone regulating phosphate and calcitriol metabolism and of klotho has changed the understanding of phosphate homeostasis; and a bone-kidney axis has emerged. In this review, we present recent findings regarding the consequences of mutations affecting several human genes encoding renal phosphate transporters or proteins regulating phosphate transport activity. We also describe the role played by the FGF23-klotho axis in phosphate homeostasis and its involvement in the pathophysiology of phosphate disturbances in chronic kidney disease.
Collapse
|
38
|
Tiosano D, Hochberg Z. Hypophosphatemia: the common denominator of all rickets. J Bone Miner Metab 2009; 27:392-401. [PMID: 19504043 DOI: 10.1007/s00774-009-0079-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 03/02/2009] [Indexed: 12/16/2022]
Abstract
Rickets is a disease of the hypertrophic chondrocytes in the growth plate and is caused by hypophosphatemia-a derived defect in terminal chondrocyte apoptosis. This highlights the critical role of phosphorous in cartilage and bone metabolism. This review shows the role of phosphorous metabolism, transport and function in maintaining phosphorous supply to the growth plate, bone osteoblast and the kidney. Given that phosphorous is the common denominator of all rickets, this review proposes a new classification for the differential diagnosis of rickets, which is based on the mechanisms leading to hypophosphatemia-high PTH activity, high FGF23 activity or renal phosphaturia.
Collapse
Affiliation(s)
- Dov Tiosano
- Meyer Children's Hospital, Rambam Medical Center, POB 9602, 31096, Haifa, Israel.
| | | |
Collapse
|
39
|
Biber J, Hernando N, Forster I, Murer H. Regulation of phosphate transport in proximal tubules. Pflugers Arch 2008; 458:39-52. [PMID: 18758808 DOI: 10.1007/s00424-008-0580-8] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Accepted: 08/13/2008] [Indexed: 12/13/2022]
Abstract
Homeostasis of inorganic phosphate (P(i)) is primarily an affair of the kidneys. Reabsorption of the bulk of filtered P(i) occurs along the renal proximal tubule and is initiated by apically localized Na(+)-dependent P(i) cotransporters. Tubular P(i) reabsorption and therefore renal excretion of P(i) is controlled by a number of hormones, including phosphatonins, and metabolic factors. In most cases, regulation of P(i) reabsorption is achieved by changing the apical abundance of Na(+)/Pi cotransporters. The regulatory mechanisms involve various signaling pathways and a number of proteins that interact with Na(+)/P(i) cotransporters.
Collapse
Affiliation(s)
- J Biber
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland.
| | | | | | | |
Collapse
|
40
|
Chaudhry FA, Boulland JL, Jenstad M, Bredahl MKL, Edwards RH. Pharmacology of neurotransmitter transport into secretory vesicles. Handb Exp Pharmacol 2008:77-106. [PMID: 18064412 DOI: 10.1007/978-3-540-74805-2_4] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Many neuropsychiatric disorders appear to involve a disturbance of chemical neurotransmission, and the mechanism of available therapeutic agents supports this impression. Postsynaptic receptors have received considerable attention as drug targets, but some of the most successful agents influence presynaptic processes, in particular neurotransmitter reuptake. The pharmacological potential of many other presynaptic elements, and in particular the machinery responsible for loading transmitter into vesicles, has received only limited attention. The similarity of vesicular transporters to bacterial drug resistance proteins and the increasing evidence for regulation of vesicle filling and recycling suggest that the pharmacological potential of vesicular transporters has been underestimated. In this review, we discuss the pharmacological effects of psychostimulants and therapeutic agents on transmitter release.
Collapse
Affiliation(s)
- Farrukh A Chaudhry
- The Biotechnology Centre of Oslo, University of Oslo, 1125, Blindern, Oslo, 0317, Norway.
| | | | | | | | | |
Collapse
|
41
|
Guo B, Jin Y, Wussler C, Blancaflor EB, Motes CM, Versaw WK. Functional analysis of the Arabidopsis PHT4 family of intracellular phosphate transporters. THE NEW PHYTOLOGIST 2008; 177:889-898. [PMID: 18086223 DOI: 10.1111/j.1469-8137.2007.02331.x] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The transport of phosphate (Pi) between subcellular compartments is central to metabolic regulation. Although some of the transporters involved in controlling the intracellular distribution of Pi have been identified in plants, others are predicted from genetic, biochemical and bioinformatics studies. Heterologous expression in yeast, and gene expression and localization in plants were used to characterize all six members of an Arabidopsis thaliana membrane transporter family designated here as PHT4. PHT4 proteins share similarity with SLC17/type I Pi transporters, a diverse group of animal proteins involved in the transport of Pi, organic anions and chloride. All of the PHT4 proteins mediate Pi transport in yeast with high specificity. Bioinformatic analysis and localization of PHT4-GFP fusion proteins indicate that five of the proteins are targeted to the plastid envelope, and the sixth resides in the Golgi apparatus. PHT4 genes are expressed in both roots and leaves, although two of the genes are expressed predominantly in leaves and one mostly in roots. These expression patterns, together with Pi transport activities and subcellular locations, suggest roles for PHT4 proteins in the transport of Pi between the cytosol and chloroplasts, heterotrophic plastids and the Golgi apparatus.
Collapse
Affiliation(s)
| | | | | | - E B Blancaflor
- Plant Biology Division, Samuel Roberts Noble Foundation, Ardmore, OK 73401, USA
| | - C M Motes
- Plant Biology Division, Samuel Roberts Noble Foundation, Ardmore, OK 73401, USA
| | - W K Versaw
- Department of Biology
- Molecular and Environmental Plant Sciences Program, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
42
|
Abstract
Changes in the response to release of a single synaptic vesicle have generally been attributed to postsynaptic modification of receptor sensitivity, but considerable evidence now demonstrates that alterations in vesicle filling also contribute to changes in quantal size. Receptors are not saturated at many synapses, and changes in the amount of transmitter per vesicle contribute to the physiological regulation of release. On the other hand, the presynaptic factors that determine quantal size remain poorly understood. Aside from regulation of the fusion pore, these mechanisms fall into two general categories: those that affect the accumulation of transmitter inside a vesicle and those that affect vesicle size. This review will summarize current understanding of the neurotransmitter cycle and indicate basic, unanswered questions about the presynaptic regulation of quantal size.
Collapse
Affiliation(s)
- Robert H Edwards
- Department of Neurology and Physiology, UCSF School of Medicine, San Francisco, CA 94158-2517, USA.
| |
Collapse
|
43
|
Virkki LV, Biber J, Murer H, Forster IC. Phosphate transporters: a tale of two solute carrier families. Am J Physiol Renal Physiol 2007; 293:F643-54. [PMID: 17581921 DOI: 10.1152/ajprenal.00228.2007] [Citation(s) in RCA: 164] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Phosphate is an essential component of life and must be actively transported into cells against its electrochemical gradient. In vertebrates, two unrelated families of Na+ -dependent P(i) transporters carry out this task. Remarkably, the two families transport different P(i) species: whereas type II Na+/P(i) cotransporters (SCL34) prefer divalent HPO(4)(2-), type III Na(+)/P(i) cotransporters (SLC20) transport monovalent H2PO(4)(-). The SCL34 family comprises both electrogenic and electroneutral members that are expressed in various epithelia and other polarized cells. Through regulated activity in apical membranes of the gut and kidney, they maintain body P(i) homeostasis, and in salivary and mammary glands, liver, and testes they play a role in modulating the P(i) content of luminal fluids. The two SLC20 family members PiT-1 and PiT-2 are electrogenic and ubiquitously expressed and may serve a housekeeping role for cell P(i) homeostasis; however, also more specific roles are emerging for these transporters in, for example, bone mineralization. In this review, we focus on recent advances in the characterization of the transport kinetics, structure-function relationships, and physiological implications of having two distinct Na+/P(i) cotransporter families.
Collapse
Affiliation(s)
- Leila V Virkki
- Institute of Physiology and Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | | | | | | |
Collapse
|
44
|
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: 10.8] [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
|
45
|
Yan F, Angel R, Ashwell CM. Characterization of the Chicken Small Intestine Type IIb Sodium Phosphate Cotransporter. Poult Sci 2007; 86:67-76. [PMID: 17179418 DOI: 10.1093/ps/86.1.67] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Intestinal absorption and renal resorption play a critical role in overall phosphorus homeostasis in chickens. Using RNase-ligase-mediated rapid amplification of cDNA ends PCR, we obtained a cDNA from the broiler small intestine that encodes a type IIb Na-dependent phosphate transporter. The cDNA has an open reading frame of 2,022 bp and predicts a 674-amino acid protein with a molecular mass of approximately 74 kDa. Prediction of membrane spanning domains based on the hydrophilic and hydrophobic properties of the amino acids suggests 8 transmembrane domains, with both the NH(2) and COOH termini being intracellular. The Na-inorganic phosphate (Pi) IIb cotransporter has relative high homology with other type II Na-Pi cotransporters but low homology with the type I or type III Na-Pi cotransporters. Northern blot analysis demonstrated the presence of a single mRNA transcript present predominantly in the small intestine, with the highest expression in the duodenum, followed by the jejunum and ileum. In situ hybridization indicated that the Na-Pi cotransporter mRNA is expressed throughout the vertical cryptvillus axis of the small intestine. Reduction of P in the diet of chicks from hatch to 4 d of age resulted in a significant induction of Na-Pi cotransporter mRNA expression in the small intestine. Further study is needed to elucidate its physiological role in intestinal phosphate absorption in chickens.
Collapse
Affiliation(s)
- F Yan
- Department of Animal and Avian Sciences, University of Maryland, College Park 20742, USA
| | | | | |
Collapse
|
46
|
Abstract
The expression of vesicular glutamate transporters (VGLUTs) 1 and 2 accounts for the ability of most traditionally accepted excitatory neurons to release glutamate by exocytosis. However, several cell populations (serotonin and dopamine neurons) have been demonstrated to release glutamate in vitro and do not obviously express these transporters. Rather, these neurons express a novel, third isoform that in fact appears confined to neurons generally associated with a transmitter other than glutamate. They include serotonin and possibly dopamine neurons, cholinergic interneurons in the striatum, and GABAergic interneurons of the hippocampus and cortex. Although the physiological role of VGLUT3 remains largely conjectural, several observations in vivo suggest that the glutamate release mediated by VGLUT3 has an important role in synaptic transmission, plasticity, and development.
Collapse
Affiliation(s)
- R P Seal
- Department of Neurology, UCSF School of Medicine, 600 16th St., GH-N272B, San Francisco CA, 94143-2140, USA
| | | |
Collapse
|
47
|
Launay-Vacher V, Izzedine H, Karie S, Hulot JS, Baumelou A, Deray G. Renal Tubular Drug Transporters. ACTA ACUST UNITED AC 2006; 103:p97-106. [PMID: 16554667 DOI: 10.1159/000092212] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2004] [Accepted: 11/03/2005] [Indexed: 01/25/2023]
Abstract
The kidney plays an important role in the elimination of numerous hydrophilic xenobiotics, including drugs, toxins, and endogenous compounds. It has developed high-capacity transport systems to prevent urinary loss of filtered nutrients, as well as electrolytes, and simultaneously to facilitate tubular secretion of a wide range of organic ions. Transport systems for organic anions and cations are primarily involved in the secretion of drugs in renal tubules. The identification and characterization of organic anion and cation transporters have been progressing at the molecular level. To date, many members of the organic anion transporter, organic cation transporter, and organic anion-transporting polypeptide families have been found to mediate the transport of diverse organic ions. It has also been suggested that ATP-dependent primary active transporters such as MDR1/P-glycoprotein and the multidrug resistance-associated protein family function as efflux pumps of renal tubular cells for more hydrophobic molecules and anionic conjugates. Tubular reabsorption of peptide-like drugs such as beta-lactam antibiotics across the brush-border membranes appears to be mediated by two distinct H+/peptide cotransporters: PEPT1 and PEPT2. Renal disposition of drugs occurs through interaction with these diverse secretory and absorptive transporters in renal tubules. Studies of the functional characteristics, such as substrate specificity and transport mechanisms, and of the localization of drug transporters could provide information regarding the cellular network involved in renal handling of drugs. Detailed information concerning molecular and cellular aspects of drug transporters expressed in the kidney has facilitated studies of the mechanisms underlying renal disposition as well as transporter-mediated drug interactions.
Collapse
|
48
|
Dudas PL, Pelis RM, Braun EJ, Renfro JL. Transepithelial urate transport by avian renal proximal tubule epithelium in primary culture. ACTA ACUST UNITED AC 2006; 208:4305-15. [PMID: 16272253 DOI: 10.1242/jeb.01879] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Birds are uricotelic, and because they excrete urate by renal tubular secretion, they provide a convenient model for examination of this process. Primary monolayer cultures of the isolated renal proximal tubule epithelium from the domestic chicken, Gallus gallus L., were mounted in Ussing chambers where several substrates/inhibitors of renal organic anion transporters were tested for the sidedness and specificity of their effects on transepithelial urate transport. Transepithelial electrical resistance, electrical potential and sodium-dependent glucose current were monitored to detect nonspecific effects. Under control short-circuited conditions the ratio of unidirectional fluxes of [(14)C]urate was found to be 3:1. Active net secretion was specifically inhibited by 1 mmol l(-1) probenecid and 10 mmol l(-1) para-aminohippuric acid (PAH). Bromocresol Green, cimetidine, nocodozole, cytochalasin D and ouabain also inhibited secretion but were toxic. Interstitial-side lithium (5 mmol l(-1)) and glutarate (1 mmol l(-1)) specifically blocked transport, but 10-100 micromol l(-1) glutarate had no effect. Interstitial estrone sulfate (ES) stimulated urate secretion at 10 micromol l(-1) but was inhibitory at 500 micromol l(-1). Active PAH secretion (5:1 flux ratio) was inhibited 34% by 330 micromol l(-1) urate. ES (500 micromol l(-1)) blocked the remainder. From the lumen side, glucose-free, Cl(-)-free and high K(+) (30 mmol l(-1)) solutions, or an alkaline pH of 7.7 had no effect on urate transport and neither did several compounds known to be uricosuric. Lumen-side methotrexate (500 micromol l(-1)) and MK571 (20 micromol l(-1)) strongly inhibited urate secretion. MK571 had no effect from the interstitial side. RT-PCR revealed mRNA for OAT1-, OAT3-, MRP2- and MRP4-like organic anion transporters in chicken proximal epithelium.
Collapse
Affiliation(s)
- Paul L Dudas
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269, USA
| | | | | | | |
Collapse
|
49
|
Prié D, Beck L, Silve C, Friedlander G. Hypophosphatemia and calcium nephrolithiasis. Nephron Clin Pract 2006; 98:e50-4. [PMID: 15499207 DOI: 10.1159/000080256] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Our knowledge of phosphate balance under physiological and pathological situations has increased substantially during the last decade thanks to the molecular identification of three dissimilar families of sodium-phosphate cotransport systems, two of them almost exclusively expressed in epithelia whereas the third one has a ubiquitous expression. Intracellular proteins such as NHERF1 (sodium-proton exchanger regulatory factor 1) can interact with phosphate transporters through PDZ domains thus regulating the expression of the transporters at the membrane. Moreover, newly acknowledged paracrine/endocrine peptides, such as fibroblast growth factor 23 (FGF23), also affect the activity of phosphate transporters. Renal phosphate leak, related to invalidation (in the mouse) or to mutations (in humans) of the renal phosphate transporter NPT2a, leads to hypophosphatemia on the one hand, and to nephrolithiasis or bone demineralization on the other hand. Similar features are observed during invalidation of NHERF or in case of overproduction of FGF23. These observations highlight the importance of phosphate homeostasis in common diseases such as renal stones or bone loss.
Collapse
Affiliation(s)
- Dominique Prié
- Department of Physiology and Inserm U 426, Faculté de Médecine Xavier Bichat, Université Denis Diderot, Paris, France
| | | | | | | |
Collapse
|
50
|
Sekine T, Miyazaki H, Endou H. Molecular physiology of renal organic anion transporters. Am J Physiol Renal Physiol 2006; 290:F251-61. [PMID: 16403838 DOI: 10.1152/ajprenal.00439.2004] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Recent advances in molecular biology have identified three organic anion transporter families: the organic anion transporter (OAT) family encoded by SLC22A, the organic anion transporting peptide (OATP) family encoded by SLC21A (SLCO), and the multidrug resistance-associated protein (MRP) family encoded by ABCC. These families play critical roles in the transepithelial transport of organic anions in the kidneys as well as in other tissues such as the liver and brain. Among these families, the OAT family plays the central role in renal organic anion transport. Knowledge of these three families at the molecular level, such as substrate selectivity, tissue distribution, and gene localization, is rapidly increasing. In this review, we will give an overview of molecular information on renal organic anion transporters and describe recent topics such as the regulatory mechanisms and molecular physiology of urate transport. We will also discuss the physiological roles of each organic anion transporter in the light of the transepithelial transport of organic anions in the kidneys.
Collapse
Affiliation(s)
- Takashi Sekine
- Kyorin University School of Medicine, Department of Pharmacology and Toxicology, 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | | | | |
Collapse
|