1
|
Are ENT1/ENT1, NOTCH3, and miR-21 Reliable Prognostic Biomarkers in Patients with Resected Pancreatic Adenocarcinoma Treated with Adjuvant Gemcitabine Monotherapy? Cancers (Basel) 2019; 11:cancers11111621. [PMID: 31652721 PMCID: PMC6893654 DOI: 10.3390/cancers11111621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 12/20/2022] Open
Abstract
Evidence on equilibrative nucleoside transporter 1 (ENT1) and microRNA-21 (miR‑21) is not yet sufficiently convincing to consider them as prognostic biomarkers for patients with pancreatic ductal adenocarcinoma (PDAC). Here, we investigated the prognostic value of ENT1/ENT1, miR-21, and neurogenic locus homolog protein 3 gene (NOTCH3) in a well-defined cohort of resected patients treated with adjuvant gemcitabine chemotherapy (n = 69). Using a combination of gene expression quantification in microdissected tissue, immunohistochemistry, and univariate/multivariate statistical analyses we did not confirm association of ENT1/ENT1 and NOTCH3 with improved disease-specific survival (DSS). Low miR-21 was associated with longer DSS in patients with negative regional lymph nodes or primary tumor at stage 1 and 2. In addition, downregulation of ENT1 was observed in PDAC of patients with high ENT1 expression in normal pancreas, whereas NOTCH3 was upregulated in PDAC of patients with low NOTCH3 levels in normal pancreas. Tumor miR‑21 was upregulated irrespective of its expression in normal pancreas. Our data confirmed that patient stratification based on expression of ENT1/ENT1 or miR‑21 is not ready to be implemented into clinical decision-making processes. We also conclude that occurrence of ENT1 and NOTCH3 deregulation in PDAC is dependent on their expression in normal pancreas.
Collapse
|
2
|
Jiraskova L, Cerveny L, Karbanova S, Ptackova Z, Staud F. Expression of Concentrative Nucleoside Transporters ( SLC28A) in the Human Placenta: Effects of Gestation Age and Prototype Differentiation-Affecting Agents. Mol Pharm 2018; 15:2732-2741. [PMID: 29782174 DOI: 10.1021/acs.molpharmaceut.8b00238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Equilibrative ( SLC29A) and concentrative ( SLC28A) nucleoside transporters contribute to proper placental development and mediate uptake of nucleosides/nucleoside-derived drugs. We analyzed placental expression of SLC28A mRNA during gestation. Moreover, we studied in choriocarcinoma-derived BeWo cells whether SLC29A and SLC28A mRNA levels can be modulated by activity of adenylyl cyclase, retinoic acid receptor activation, CpG islands methylation, or histone acetylation, using forskolin, all- trans-retinoic acid, 5-azacytidine, and sodium butyrate/sodium valproate, respectively. We found that expression of SLC28A1, SLC28A2, and SLC28A3 increases during gestation and reveals considerable interindividual variability. SLC28A2 was shown to be a dominant subtype in the first-trimester and term human placenta, while SLC28A1 exhibited negligible expression in the term placenta only. In BeWo cells, we detected mRNA of SLC28A2 and SLC28A3. Levels of the latter were affected by 5-azacytidine and all- trans-retinoic acid, while the former was modulated by sodium valproate (but not sodium butyrate), all- trans-retinoic acid, 5-azacytidine, and forskolin that caused 25-fold increase in SLC28A2 mRNA; we documented by analysis of syncytin-1 that the observed changes in SLC28A expression do not correlate with the morphological differentiation state of BeWo cells. Upregulated SLC28A2 mRNA was reflected in elevated uptake of [3H]-adenosine, high-affinity substrate of concentrative nucleoside transporter 2. Using KT-5720 and inhibitors of phosphodiesterases, we subsequently confirmed importance of cAMP/protein kinase A pathway in SLC28A2 regulation. On the other hand, SLC29A genes exhibited constitutive expression and none of the tested compounds increased SLC28A1 expression to detectable levels. In conclusion, we provide the first evidence that methylation status and activation of retinoic acid receptor affect placental SLC28A2 and SLC28A3 transcription and substrates of concentrative nucleoside transporter 2 might be taken up in higher extent in placentas with overactivated cAMP/protein kinase A pathway and likely in the term placenta.
Collapse
Affiliation(s)
- Lucie Jiraskova
- Department of Pharmacology and Toxicology , Charles University, Faculty of Pharmacy in Hradec Kralove , Akademika Heyrovskeho 1203 , 50005 Hradec Kralove , Czech Republic
| | - Lukas Cerveny
- Department of Pharmacology and Toxicology , Charles University, Faculty of Pharmacy in Hradec Kralove , Akademika Heyrovskeho 1203 , 50005 Hradec Kralove , Czech Republic
| | - Sara Karbanova
- Department of Pharmacology and Toxicology , Charles University, Faculty of Pharmacy in Hradec Kralove , Akademika Heyrovskeho 1203 , 50005 Hradec Kralove , Czech Republic
| | - Zuzana Ptackova
- Department of Pharmacology and Toxicology , Charles University, Faculty of Pharmacy in Hradec Kralove , Akademika Heyrovskeho 1203 , 50005 Hradec Kralove , Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology , Charles University, Faculty of Pharmacy in Hradec Kralove , Akademika Heyrovskeho 1203 , 50005 Hradec Kralove , Czech Republic
| |
Collapse
|
3
|
Urtasun N, Boces-Pascual C, Boix L, Bruix J, Pastor-Anglada M, Pérez-Torras S. Role of drug-dependent transporter modulation on the chemosensitivity of cholangiocarcinoma. Oncotarget 2017; 8:90185-90196. [PMID: 29163820 PMCID: PMC5685741 DOI: 10.18632/oncotarget.21624] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 08/27/2017] [Indexed: 12/22/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a heterogeneous group of malignancies with limited therapeutic options. Curative therapy is limited to surgery whereas chemotherapy treatments are the election option for unresectable or metastatic cholangiocarcinoma. Cisplatin plus gemcitabine is the reference chemotherapy regimen, albeit the contribution to the median overall survival barely reaches one year. Drug transporters are undoubtedly a limiting step for drug bioavailability and have been clearly related to chemoresistance. Several members of the SoLute Carrier (SLC) superfamily involved in the uptake of anticancer drugs used to treat cholangiocarcinoma are downregulated in these tumors. This study shows the increase in the expression of specific drug transporters exerted by cisplatin treatment thereby enhancing their transport activity. Combination treatments of cisplatin with selected drugs as gemcitabine and sorafenib take in by these transporters at the desired combination schedule induced synergy. These data support the concept that proper administration pattern could favor treatment outcome.
Collapse
Affiliation(s)
- Nerea Urtasun
- Molecular Pharmacology and Experimental Therapeutics (MPET), Section Biochemistry and Molecular Pharmacology, Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain.,Oncology Program, CIBER ehd, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Clara Boces-Pascual
- Molecular Pharmacology and Experimental Therapeutics (MPET), Section Biochemistry and Molecular Pharmacology, Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain
| | - Loreto Boix
- Oncology Program, CIBER ehd, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain.,Barcelona Clinic Liver Cancer (BCLC) Group, Liver Unit, Hospital Clínic of Barcelona, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica (FCRB), Barcelona, Spain
| | - Jordi Bruix
- Oncology Program, CIBER ehd, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain.,Barcelona Clinic Liver Cancer (BCLC) Group, Liver Unit, Hospital Clínic of Barcelona, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica (FCRB), Barcelona, Spain
| | - Marçal Pastor-Anglada
- Molecular Pharmacology and Experimental Therapeutics (MPET), Section Biochemistry and Molecular Pharmacology, Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain.,Oncology Program, CIBER ehd, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Sandra Pérez-Torras
- Molecular Pharmacology and Experimental Therapeutics (MPET), Section Biochemistry and Molecular Pharmacology, Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain.,Oncology Program, CIBER ehd, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
4
|
Grañé-Boladeras N, Pérez-Torras S, Lozano JJ, Romero MR, Mazo A, Marín JJ, Pastor-Anglada M. Pharmacogenomic analyzis of the responsiveness of gastrointestinal tumor cell lines to drug therapy: A transportome approach. Pharmacol Res 2016; 113:364-375. [DOI: 10.1016/j.phrs.2016.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/17/2016] [Accepted: 09/07/2016] [Indexed: 01/20/2023]
|
5
|
Arimany-Nardi C, Errasti-Murugarren E, Minuesa G, Martinez-Picado J, Gorboulev V, Koepsell H, Pastor-Anglada M. Nucleoside transporters and human organic cation transporter 1 determine the cellular handling of DNA-methyltransferase inhibitors. Br J Pharmacol 2015; 171:3868-80. [PMID: 24780098 DOI: 10.1111/bph.12748] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 04/02/2014] [Accepted: 04/15/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Inhibitors of DNA methyltransferases (DNMTs), such as azacytidine, decitabine and zebularine, are used for the epigenetic treatment of cancer. Their action may depend upon their translocation across the plasma membrane. The aim of this study was to identify transporter proteins contributing to DNMT inhibitor action. EXPERIMENTAL APPROACH Drug interactions with selected hCNT and hENT proteins were studied in transiently transfected HeLa and MDCK cells. Interaction with human organic cation transporters (hOCTs) was assessed in transiently transfected HeLa cells and Xenopus laevis oocytes. KEY RESULTS Zebularine uptake was mediated by hCNT1, hCNT3 and hENT2. Decitabine interacted with but was not translocated by any nucleoside transporter (NT) type. hCNT expression at the apical domain of MDCK cells promoted net vectorial flux of zebularine. Neither hOCT1 nor hOCT2 transported decitabine, but both were involved in the efflux of zebularine, suggesting these proteins act as efflux transporters. hOCT1 polymorphic variants, known to alter function, decreased zebularine efflux. CONCLUSIONS AND IMPLICATIONS This study highlights the influence of human NTs and hOCTs on the pharmacokinetics and pharmacodynamics of selected DNMT inhibitors. As hOCTs may also behave as efflux transporters, they could contribute either to chemoresistance or to chemosensitivity, depending upon the nature of the drug or combination of drugs being used in cancer therapy.
Collapse
Affiliation(s)
- C Arimany-Nardi
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona (IBUB) & National Biomedical Research Institute on Liver and Gastrointestinal Diseaes (CIBERehd), Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
6
|
Pastor-Anglada M, Pérez-Torras S. Nucleoside transporter proteins as biomarkers of drug responsiveness and drug targets. Front Pharmacol 2015; 6:13. [PMID: 25713533 PMCID: PMC4322540 DOI: 10.3389/fphar.2015.00013] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/13/2015] [Indexed: 12/13/2022] Open
Abstract
Nucleoside and nucleobase analogs are currently used in the treatment of solid tumors, lymphoproliferative diseases, viral infections such as hepatitis and AIDS, and some inflammatory diseases such as Crohn. Two gene families are implicated in the uptake of nucleosides and nucleoside analogs into cells, SCL28 and SLC29. The former encodes hCNT1, hCNT2, and hCNT3 proteins. They translocate nucleosides in a Na+ coupled manner with high affinity and some substrate selectivity, being hCNT1 and hCNT2 pyrimidine- and purine-preferring, respectively, and hCNT3 a broad selectivity transporter. SLC29 genes encode four members, being hENT1 and hENT2 the only two which are unequivocally implicated in the translocation of nucleosides and nucleobases (the latter mostly via hENT2) at the cell plasma membrane. Some nucleoside-derived drugs can also interact with and be translocated by members of the SLC22 gene family, particularly hOCT and hOAT proteins. Inter-individual differences in transporter function and perhaps, more importantly, altered expression associated with the disease itself might modulate the transporter profile of target cells, thereby determining drug bioavailability and action. Drug transporter pharmacology has been periodically reviewed. Thus, with this contribution we aim at providing a state-of-the-art overview of the clinical evidence generated so far supporting the concept that these membrane proteins can indeed be biomarkers suitable for diagnosis and/or prognosis. Last but not least, some of these transporter proteins can also be envisaged as drug targets, as long as they can show “transceptor” functions, in some cases related to their role as modulators of extracellular adenosine levels, thereby providing a functional link between P1 receptors and transporters.
Collapse
Affiliation(s)
- Marçal Pastor-Anglada
- Molecular Pharmacology and Experimental Therapeutics, Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University of Barcelona, Barcelona Spain ; Oncology Program, CIBER ehd, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona Spain
| | - Sandra Pérez-Torras
- Molecular Pharmacology and Experimental Therapeutics, Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University of Barcelona, Barcelona Spain ; Oncology Program, CIBER ehd, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona Spain
| |
Collapse
|
7
|
Arimany-Nardi C, Montraveta A, Lee-Vergés E, Puente XS, Koepsell H, Campo E, Colomer D, Pastor-Anglada M. Human organic cation transporter 1 (hOCT1) as a mediator of bendamustine uptake and cytotoxicity in chronic lymphocytic leukemia (CLL) cells. THE PHARMACOGENOMICS JOURNAL 2015; 15:363-71. [PMID: 25582574 DOI: 10.1038/tpj.2014.77] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/13/2014] [Accepted: 11/05/2014] [Indexed: 01/20/2023]
Abstract
Bendamustine is used in the treatment of chronic lymphocytic leukemia (CLL). Routes for bendamustine entry into target cells are unknown. This study aimed at identifying transporter proteins implicated in bendamustine uptake. Our results showed that hOCT1 is a bendamustine transporter, as bendamustine could cis-inhibit the uptake of a canonical hOCT1 substrate, with a Ki in the micromolar range, consistent with the EC50 values of the cytotoxicity triggered by this drug in HEK293 cells expressing hOCT1. hOCT1 polymorphic variants determining impaired bendamustine-transporter interaction, consistently reduced bendamustine cytotoxicity in HEK293 cells stably expressing them. Exome genotyping of the SLC22A1 gene, encoding hOCT1, was undertaken in a cohort of 241 CLL patients. Ex vivo cytotoxicity to bendamustine was measured in a subset of cases and shown to correlate with SLC22A1 polymorphic variants. In conclusion, hOCT1 is a suitable bendamustine transporter, thereby contributing to its cytotoxic effect depending upon the hOCT1 genetic variants expressed.
Collapse
Affiliation(s)
- C Arimany-Nardi
- 1] Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University of Barcelona, Barcelona, Spain [2] Oncology Program, National Biomedical Research Institute of Liver and Gastrointestinal Diseases (CIBER ehd), Instituto de Salud Carlos III, Madrid, Spain
| | - A Montraveta
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - E Lee-Vergés
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - X S Puente
- Institute of Oncology, University of Oviedo, Oviedo, Spain
| | - H Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Insitute, University of Würzburg, Würzburg, Germany
| | - E Campo
- Hematopathology Unit, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - D Colomer
- 1] Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain [2] Hematopathology Unit, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - M Pastor-Anglada
- 1] Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University of Barcelona, Barcelona, Spain [2] Oncology Program, National Biomedical Research Institute of Liver and Gastrointestinal Diseases (CIBER ehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Godoy V, Banales JM, Medina JF, Pastor-Anglada M. Functional crosstalk between the adenosine transporter CNT3 and purinergic receptors in the biliary epithelia. J Hepatol 2014; 61:1337-43. [PMID: 25034758 DOI: 10.1016/j.jhep.2014.06.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 06/17/2014] [Accepted: 06/27/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Both hepatocytes and cholangiocytes release ATP into the bile, where it acts as a potent autocrine/paracrine stimulus that activates biliary secretory mechanisms. ATP is known to be metabolized into multiple breakdown products, ultimately yielding adenosine. However, the elements implicated in the adenosine-dependent purinergic regulation of cholangiocytes are not known. METHODS Normal rat cholangiocytes (NRCs) were used to study the expression of adenosine receptors and transporters and their functional interactions at the apical and basolateral membrane domains of polarized cholangiocytes. RESULTS We found that: (1) cholangiocytes exclusively express two concentrative nucleoside transporters (CNT) known to be efficient adenosine carriers: CNT3, located at the apical membrane, and CNT2, located at apical and basolateral membrane domains; (2) in both domains, NRCs also express the high affinity adenosine receptor A2A, which modulated the activity of apical CNT3 in a domain-specific manner; (3) the regulation exerted by A2A on CNT3 was dependent upon the cAMP/PKA/ERK/CREB axis, intracellular trafficking mechanisms and AMPK phosphorylation; (4) secretin increased the activity of the apically-located CNT3, and promoted additional basolateral CNT3-related activity; and (5) extracellular ATP (a precursor of adenosine) was able to exert an inhibitory effect on the apical activity of both CNT3 and CNT2. CONCLUSIONS This study uncovered the functional expression of nucleoside transporters in cholangiocytes and provides evidence for direct crosstalks between adenosine transporters and receptors for adenosine and its natural extracellular precursor, ATP. Our data anticipate the possibility of adenosine playing a major role in the physiopathology of the biliary epithelia.
Collapse
Affiliation(s)
- Valeria Godoy
- Molecular Pharmacology and Experimental Therapeutics, Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Spain
| | - Jesús M Banales
- Department of Liver Diseases, Biodonostia Research Institute (Donostia University Hospital), IKERBASQUE (Basque Foundation for Science), University of Basque Country (UPV/EHU), San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Spain
| | - Juan F Medina
- Molecular Genetics, Division of Gene Therapy and Hepatology, School of Medicine and CIMA of the University of Navarra, Pamplona, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Spain
| | - Marçal Pastor-Anglada
- Molecular Pharmacology and Experimental Therapeutics, Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Spain.
| |
Collapse
|
9
|
Cusato J, Allegra S, De Nicolò A, Boglione L, Fatiguso G, Cariti G, Ciancio A, Smedile A, Strona S, Troshina G, Rizzetto M, Di Perri G, D'Avolio A. ABCB11 and ABCB1 gene polymorphisms impact on telaprevir pharmacokinetic at one month of therapy. Biomed Pharmacother 2014; 69:63-9. [PMID: 25661339 DOI: 10.1016/j.biopha.2014.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/05/2014] [Indexed: 12/24/2022] Open
Abstract
In 2011 direct-acting antivirals, including telaprevir, have been developed to achieve a better antiviral effect. It was reported that telaprevir is a substrate of P-glycoprotein (ABCB1) and cytochrome P450 3A4. The aim of this retrospective study was the evaluation of the influence of some single nucleotide polymorphisms (SNPs) of genes (ABCB1, SLC28A2/3, SLC29A1) involved in TLV and RBV transport and their correlation with plasma TLV drug exposure at 1 month of therapy. We also investigated the association of a SNP in ABCB11 gene, whose role in TLV transport was not yet shown. Twenty-nine HCV-1 patients treated with telaprevir, ribavirin and pegylated-interferon-α were retrospectively analyzed; allelic discrimination was performed by real-time PCR. Telaprevir Ctrough levels were influenced by Metavir score (P=0.023), ABCB1 2677 G>T (P=0.006), ABCB1 1236 C>T (P=0.015) and ABCB11 1131 T>C (P=0.033) SNPs. Regarding ABCB1 3435 C>T, a not statistically significant trend in telaprevir plasma concentration was observed. Metavir score (P=0.002, OR -336; 95% CI -535;-138), ABCB1 2677 (P=0.020, OR 497; 95% CI 86; 910), ABCB11 1131 (P=0.002, OR 641; 95% CI 259;1023) and CNT2 -146 (P=0.006, OR -426; 95% CI -721;-132) were able to predict telaprevir plasma levels in the regression analysis. Other SNPs showed no association. This study reveals BSEP implication in telaprevir transport and confirms the involvement and influence of P-glycoprotein on telaprevir plasma levels. To date, no similar data concerning pharmacogenetics and pharmacokinetics were published, but further studies in different and bigger cohorts are needed.
Collapse
Affiliation(s)
- Jessica Cusato
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy.
| | - Sarah Allegra
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - Amedeo De Nicolò
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - Lucio Boglione
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - Giovanna Fatiguso
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - Giuseppe Cariti
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - Alessia Ciancio
- Unit of Gastroenterology, University of Turin, Department of Medical Sciences, S. Giovanni Battista (Molinette) Hospital, Turin, Italy
| | - Antonina Smedile
- Unit of Gastroenterology, University of Turin, Department of Medical Sciences, S. Giovanni Battista (Molinette) Hospital, Turin, Italy
| | - Silvia Strona
- Unit of Gastroenterology, University of Turin, Department of Medical Sciences, S. Giovanni Battista (Molinette) Hospital, Turin, Italy
| | - Giulia Troshina
- Unit of Gastroenterology, University of Turin, Department of Medical Sciences, S. Giovanni Battista (Molinette) Hospital, Turin, Italy
| | - Mario Rizzetto
- Unit of Gastroenterology, University of Turin, Department of Medical Sciences, S. Giovanni Battista (Molinette) Hospital, Turin, Italy
| | - Giovanni Di Perri
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| | - Antonio D'Avolio
- Unit of Infectious Diseases, University of Turin, Department of Medical Sciences, Amedeo di Savoia Hospital, Turin, Italy
| |
Collapse
|
10
|
rCNT2 extracellular cysteines, Cys615
and Cys649
, are important for maturation and sorting to the plasma membrane. FEBS Lett 2014; 588:4382-9. [DOI: 10.1016/j.febslet.2014.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/27/2014] [Accepted: 10/06/2014] [Indexed: 12/11/2022]
|
11
|
Pinilla-Macua I, Fernández-Calotti P, Pérez-Del-Pulgar S, Pastor-Anglada M. Ribavirin uptake into human hepatocyte HHL5 cells is enhanced by interferon-α via up-regulation of the human concentrative nucleoside transporter (hCNT2). Mol Pharm 2014; 11:3223-30. [PMID: 24957263 DOI: 10.1021/mp500263p] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ribavirin is a broad spectrum antiviral that increases the response rate in chronic hepatitis C patients when administered in combination with IFNα. Ribavirin is a purine nucleoside derivative, transported into hepatocytes by nucleoside transporters. hCNT2 is the best candidate to mediate ribavirin uptake into hepatocytes due to its high-affinity for purines and its capacity to concentrate its substrates intracellularly. The aim of this study was to determine whether hCNT2 function is under IFNα modulation. IFNα treatment of the nontransformed human hepatocyte-derived cell line HHL5 induced a rapid and transient increase in hCNT2 activity after cytokine addition. hCNT2 activity up-regulation was associated with increased ribavirin accumulation into cells. This increase was consistent with the translocation of hCNT2-containing vesicles to the plasma membrane via a mechanism requiring ERK 1/2 and ROCK activation and cytoskeleton integrity. Longer treatments with IFNα induced transcriptional activation of the hCNT2-encoding gene (SLC28A2), resulting in a sustained increase in hCNT2-related activity. These observations are proof of concept for at least one of the putative mechanisms underlying the synergistic responses induced by combination therapy with IFNα and ribavirin.
Collapse
Affiliation(s)
- Itziar Pinilla-Macua
- Department of Biochemistry and Molecular Biology, University of Barcelona, Institute of Biomedicine (IBUB) , 08028 Barcelona, Spain
| | | | | | | |
Collapse
|
12
|
Hypoxia and P1 receptor activation regulate the high-affinity concentrative adenosine transporter CNT2 in differentiated neuronal PC12 cells. Biochem J 2013; 454:437-45. [PMID: 23819782 DOI: 10.1042/bj20130231] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Under several adverse conditions, such as hypoxia or ischaemia, extracellular levels of adenosine are elevated because of increased energy demands and ATP metabolism. Because extracellular adenosine affects metabolism through G-protein-coupled receptors, its regulation is of high adaptive importance. CNT2 (concentrative nucleoside transporter 2) may play physiological roles beyond nucleoside salvage in brain as it does in other tissues. Even though nucleoside transport in brain has mostly been seen as being of equilibrative-type, in the present study, we prove that the rat phaeochromocytoma cell line PC12 shows a concentrative adenosine transport of CNT2-type when cells are differentiated to a neuronal phenotype by treatment with NGF (nerve growth factor). Differentiation of PC12 cells was also associated with the up-regulation of adenosine A1 receptors. Addition of adenosine receptor agonists to cell cultures increased CNT2-related activity by a mechanism consistent with A₁ and A2A receptor activation. The addition of adenosine to the culture medium also induced the phosphorylation of the intracellular regulatory kinase AMPK (AMP-activated protein kinase), with this effect being dependent upon adenosine transport. CNT2-related activity of differentiated PC12 cells was also dramatically down-regulated under hypoxic conditions. Interestingly, the analysis of nucleoside transporter expression after experimental focal ischaemia in rat brain showed that CNT2 expression was down-regulated in the infarcted tissue, with this effect somehow being restricted to other adenosine transporter proteins such as CNT3 and ENT1 (equilibrative nucleoside transporter 1). In summary, CNT2 is likely to modulate extracellular adenosine and cell energy balance in neuronal tissue.
Collapse
|
13
|
Zhang Z, Liu R, Townsend PA, Proud CG. p90RSKs mediate the activation of ribosomal RNA synthesis by the hypertrophic agonist phenylephrine in adult cardiomyocytes. J Mol Cell Cardiol 2013; 59:139-47. [DOI: 10.1016/j.yjmcc.2013.03.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 03/05/2013] [Accepted: 03/07/2013] [Indexed: 01/06/2023]
|
14
|
Concentrative nucleoside transporter 1 (hCNT1) promotes phenotypic changes relevant to tumor biology in a translocation-independent manner. Cell Death Dis 2013; 4:e648. [PMID: 23722537 PMCID: PMC3674379 DOI: 10.1038/cddis.2013.173] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Nucleoside transporters (NTs) mediate the uptake of nucleosides and nucleobases across the plasma membrane, mostly for salvage purposes. The canonical NTs belong to two gene families, SLC29 and SLC28. The former encode equilibrative nucleoside transporter proteins (ENTs), which mediate the facilitative diffusion of natural nucleosides with broad selectivity, whereas the latter encode concentrative nucleoside transporters (CNTs), which are sodium-coupled and show high affinity for substrates with variable selectivity. These proteins are expressed in most cell types, exhibiting apparent functional redundancy. This might indicate that CNTs have specific roles in the physiology of the cell beyond nucleoside salvage. Here, we addressed this possibility using adenoviral vectors to restore tumor cell expression of hCNT1 or a polymorphic variant (hCNT1S546P) lacking nucleoside translocation ability. We found that hCNT1 restoration in pancreatic cancer cells significantly altered cell-cycle progression and phosphorylation status of key signal-transducing kinases, promoted poly-(ADP-ribose) polymerase hyperactivation and cell death and reduced cell migration. Importantly, the translocation-defective transporter triggered these same effects on cell physiology. Moreover, this study also shows that restoration of hCNT1 expression is able to reduce tumor growth in a mouse model of pancreatic adenocarcinoma. These data predict a novel role for a NT protein, hCNT1, which appears to be independent of its role as mediator of nucleoside uptake by cells. Thereby, hCNT1 fits the profile of a transceptor in a substrate translocation-independent manner and is likely to be relevant to tumor biology.
Collapse
|
15
|
Huber-Ruano I, Errasti-Murugarren E, Godoy V, Vera Á, Andreu AL, Garcia-Arumi E, Martí R, Pastor-Anglada M. Functional outcome of a novel SLC29A3 mutation identified in a patient with H syndrome. Biochem Biophys Res Commun 2012; 428:532-7. [DOI: 10.1016/j.bbrc.2012.09.143] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 09/29/2012] [Indexed: 01/24/2023]
|
16
|
Errasti-Murugarren E, Fernández-Calotti P, Veyhl-Wichmann M, Diepold M, Pinilla-Macua I, Pérez-Torras S, Kipp H, Koepsell H, Pastor-Anglada M. Role of the transporter regulator protein (RS1) in the modulation of concentrative nucleoside transporters (CNTs) in epithelia. Mol Pharmacol 2012; 82:59-67. [PMID: 22492015 DOI: 10.1124/mol.111.076992] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
SLC28 genes encode three plasma membrane transporter proteins, human concentrative nucleoside transporter (CNT)1, CNT2, and CNT3, all of which are implicated in the uptake of natural nucleosides and a variety of nucleoside analogs used in the chemotherapy of cancer and viral and inflammatory diseases. Mechanisms determining their trafficking toward the plasma membrane are not well known, although this might eventually become a target for therapeutic intervention. The transporter regulator RS1, which was initially identified as a short-term, post-transcriptional regulator of the high-affinity, Na(+)-coupled, glucose transporter sodium-dependent glucose cotransporter 1, was evaluated in this study as a candidate for coordinate regulation of membrane insertion of human CNT-type proteins. With a combination of studies with mammalian cells, Xenopus laevis oocytes, and RS1-null mice, evidence that RS1 down-regulates the localization and activity at the plasma membrane of the three members of this protein family (CNT1, CNT2, and CNT3) is provided, which indicates the biochemical basis for coordinate regulation of nucleoside uptake ability in epithelia and probably in other RS1-expressing cell types.
Collapse
Affiliation(s)
- Ekaitz Errasti-Murugarren
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona and National Biomedical Research Institute of Liver and Gastrointestinal Diseases, Diagonal 643, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Guillén-Gómez E, Pinilla-Macua I, Pérez-Torras S, Choi DS, Arce Y, Ballarín JA, Pastor-Anglada M, Díaz-Encarnación MM. New role of the human equilibrative nucleoside transporter 1 (hENT1) in epithelial-to-mesenchymal transition in renal tubular cells. J Cell Physiol 2012; 227:1521-8. [PMID: 21678404 DOI: 10.1002/jcp.22869] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is an important pro-fibrotic event in which tubular epithelial cells are transformed into myofibroblasts. Nucleoside transporters (NT) are regulated by many factors and processes, some of which are involved in fibrosis, such as cytokines, inflammation, and proliferation. Equilibrative nucleoside transporter 1 (ENT1) has been proved to be the most widely expressed adenosine transporter. In that sense, ENT1 may be a key player in cell damage signaling. Here we analyze the role of human ENT1 (hENT1) in the EMT process in proximal tubular cells. Addition of the main inducer of EMT, the transforming growth factor-β1, to HK-2 cells increased hENT1 mRNA and protein level expression. ENT1-mediated adenosine uptake was also enhanced. When cells were incubated with dipyridamole to evaluate the potential contribution of ENT1 to EMT by blocking its transport activity, EMT was induced. Moreover, the knock down of hENT1 with siRNA induced EMT and collagen production in HK-2 cells. Kidneys isolated from ENT1 knockout mice showed higher levels of interstitial collagen and α-SMA positive cells than wild-type mice. Our results point to a new potential role of hENT1 as a modulator of EMT in proximal tubular cells. In this sense, hENT1 could be involved in renal protection processes, and the loss or reduced expression of hENT1 would lead to an increased vulnerability of cells to the onset and/or progression of renal fibrosis.
Collapse
Affiliation(s)
- Elena Guillén-Gómez
- Laboratori de Biologia Molecular, Universitat Autònoma de Barcelona, Fundació Puigvert, REDinREN, Institut Investigació Biosanitaria Sant Pau, Barcelona, Catalonia, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Structural determinants for rCNT2 sorting to the plasma membrane of polarized and non-polarized cells. Biochem J 2012; 442:517-25. [DOI: 10.1042/bj20110605] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
rCNT2 (rat concentrative nucleoside transporter 2) (Slc28a2) is a purine-preferring concentrative nucleoside transporter. It is expressed in both non-polarized and polarized cells, where it is localized in the brush border membrane. Since no information about the domains implicated in the plasma membrane sorting of rCNT2 is available, the present study aimed to identify structural and functional requirements for rCNT2 trafficking. The comprehensive topological mapping of the intracellular N-terminal tail revealed two main features: (i) a glutamate-enriched region (NPGLELME) between residues 21 and 28 that seems to be implicated in the stabilization of rCNT2 in the cell surface, since mutagenesis of these conserved glutamates resulted in enhanced endocytosis; and (ii) mutation of a potential protein kinase CK2 domain that led to a loss of brush border-specific sorting. Although the shortest proteins assayed (rCNT2-74AA, -48AA and -37AA) accumulated intracellularly and lost their brush border membrane preference, they were still functional. A deeper analysis of CK2 implication in CNT2 trafficking, using a CK2-specific inhibitor [DMAT (2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole)] and other complementary mutations mimicking the negative charge provided by phosphorylation (S46D and S46E), demonstrated an effect of this kinase on rCNT2 activity. In summary, the N-terminal tail of rCNT2 contains dual sorting signals. An acidic region is responsible for its proper stabilization at the plasma membrane, whereas the putative CK2 domain (Ser46) is implicated in the apical sorting of the transporter.
Collapse
|
19
|
Cano-Soldado P, Gorraitz E, Errasti-Murugarren E, Casado FJ, Lostao MP, Pastor-Anglada M. Functional analysis of the human concentrative nucleoside transporter-1 variant hCNT1S546P provides insight into the sodium-binding pocket. Am J Physiol Cell Physiol 2012; 302:C257-66. [DOI: 10.1152/ajpcell.00198.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
SLC28 genes, encoding concentrative nucleoside transporter proteins (CNT), show little genetic variability, although a few single nucleotide polymorphisms (SNPs) have been associated with marked functional disturbances. In particular, human CNT1S546P had been reported to result in negligible thymidine uptake. In this study we have characterized the molecular mechanisms responsible for this apparent loss of function. The hCNT1S546P variant showed an appropriate endoplasmic reticulum export and insertion into the plasma membrane, whereas loss of nucleoside translocation ability affected all tested nucleoside and nucleoside-derived drugs. Site-directed mutagenesis analysis revealed that it is the lack of the serine residue itself responsible for the loss of hCNT1 function. This serine residue is highly conserved, and mutation of the analogous serine in hCNT2 (Ser541) and hCNT3 (Ser568) resulted in total and partial loss of function, respectively. Moreover, hCNT3, the only member that shows a 2Na+/1 nucleoside stoichiometry, showed altered Na+ binding properties associated with a shift in the Hill coefficient, consistent with one Na+ binding site being affected by the mutation. Two-electrode voltage-clamp studies using the hCNT1S546P mutant revealed the occurrence of Na+ leak, which was dependent on the concentration of extracellular Na+ indicating that, although the variant is unable to transport nucleosides, there is an uncoupled sodium transport.
Collapse
Affiliation(s)
- Pedro Cano-Soldado
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona and CIBER EHD, Barcelona
| | - Edurne Gorraitz
- Department of Food Science, Physiology and Toxicology, University of Navarra, Pamplona, Spain
| | - Ekaitz Errasti-Murugarren
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona and CIBER EHD, Barcelona
| | - F. Javier Casado
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona and CIBER EHD, Barcelona
| | - M. Pilar Lostao
- Department of Food Science, Physiology and Toxicology, University of Navarra, Pamplona, Spain
| | - Marçal Pastor-Anglada
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona and CIBER EHD, Barcelona
| |
Collapse
|
20
|
Iadevaia V, Zhang Z, Jan E, Proud CG. mTOR signaling regulates the processing of pre-rRNA in human cells. Nucleic Acids Res 2011; 40:2527-39. [PMID: 22121221 PMCID: PMC3315323 DOI: 10.1093/nar/gkr1040] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Signaling through the mammalian target of rapamycin, complex 1 (mTORC1), positively regulates the transcription of ribosomal RNA (rRNA) and the synthesis of ribosomal proteins, thereby promoting the complex process of ribosome biogenesis. The major rRNAs are transcribed as a single precursor, which must be processed to create the 5.8S, 18S and 28S rRNAs. We used a new non-radioactive labeling approach to study the effects of rapamycin, an inhibitor of mTORC1, on rRNA synthesis. Rapamycin not only impaired synthesis of new 18S, 28S or 5S rRNA but also induced their decay. This prompted us to examine the effects of rapamycin on rRNA processing. We show that rapamycin also interferes with the processing events that generate 18S and 28S rRNA. rRNA transcription and processing occur in regions of the nucleus known as nucleoli. We find that the mTORC1 components raptor and mTOR are both present in nucleoli, where they may regulate rRNA maturation events. While rapamycin has no effect on overall nucleolar morphology or its proteome, it does induce loss of mTOR and raptor from them. These data show that mTORC1 is located in nucleoli where it acts to regulate events involved in ribosome biogenesis including the maturation of rRNA molecules.
Collapse
|
21
|
Plotnik DA, McLaughlin LJ, Chan J, Redmayne-Titley JN, Schwartz JL. The role of nucleoside/nucleotide transport and metabolism in the uptake and retention of 3'-fluoro-3'-deoxythymidine in human B-lymphoblast cells. Nucl Med Biol 2011; 38:979-86. [PMID: 21982569 DOI: 10.1016/j.nucmedbio.2011.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 03/13/2011] [Accepted: 03/26/2011] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Recent studies in the human adenocarcinoma cell line A549 have identified cell growth-dependent equilibrative nucleoside transporter-1 (hENT1) as a modifier of 3'-fluoro-3'-deoxythymidine (FLT) uptake and retention. In the present study, we used the ability to isolate human lymphoblastoid clones deficient in thymidine kinase 1 (TK1) to study how metabolism and nucleoside transport influence FLT uptake and retention. METHODS Transport and metabolism of FLT were measured in the human lymphoblastoid cell line TK6 and in eight clones isolated from TK6. Four clones were TK1-proficient, while four were TK1-deficient. Both influx and efflux of FLT were measured under conditions where concentrative and equilibrative transport could be distinguished. RESULTS Sodium-dependent concentrative FLT transport dominated over equilibrative transport mechanisms and while inhibition of hENT1 reduced FLT uptake, there were no correlations between clonal variations in hENT1 levels and FLT uptake. There was an absolute requirement of TK1 for concentration of FLT in TK6 cells. FLT uptake reached a peak after 60 min of incubation with FLT after which intracellular levels of FLT and FLT metabolites declined. Efflux was rapid and was associated with reductions in FLT and each of its metabolites. Both FLT and FLT-monophosphate were found in the efflux buffer. CONCLUSIONS Initial rates of FLT uptake were a function of both concentrative and equilibrative transporters. TK1 activity was an absolute requirement for the accumulation of FLT. Retention was dependent on nucleoside/nucleotide efflux and retrograde metabolism of FLT nucleotides.
Collapse
Affiliation(s)
- David A Plotnik
- Department of Radiation Oncology, University of Washington, Box 356069, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
22
|
Errasti-Murugarren E, Casado FJ, Pastor-Anglada M. Different N-terminal motifs determine plasma membrane targeting of the human concentrative nucleoside transporter 3 in polarized and nonpolarized cells. Mol Pharmacol 2010; 78:795-803. [PMID: 20643903 DOI: 10.1124/mol.110.065920] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Human concentrative nucleoside transporter 3 (hCNT3) is a broad-selectivity, high-affinity protein implicated in the uptake of most nucleoside-derived anticancer and antiviral drugs. Regulated trafficking of hCNT3 has been recently postulated as a suitable way to improve nucleoside-based therapies. Moreover, the recent identification of a putative novel hCNT3-type transporter lacking the first 69 amino acids and retained at the endoplasmic reticulum anticipated that the N terminus of hCNT3 contains critical motifs implicated in trafficking. In the current study, we have addressed this issue by using deletions and site-directed mutagenesis and plasma membrane expression and nucleoside uptake kinetic analysis. Data reveal that 1) a segment between amino acids 50 and 62 contains plasma membrane-sorting determinants in nonpolarized cells; 2) in particular, the Val(57)-Thr(58)-Val(59) tripeptide seems to be the core of the export signal, whereas acidic motifs upstream and downstream of it seem to be important for the kinetics of the process; and 3) in polarized epithelia, the β-turn-forming motif (17)VGFQ(20) is necessary for proper apical expression of the protein.
Collapse
Affiliation(s)
- Ekaitz Errasti-Murugarren
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona and CIBER EHD, Avda Diagonal 645, Edifici annex, Planta-1, 08028 Barcelona, Spain
| | | | | |
Collapse
|
23
|
Huber-Ruano I, Pinilla-Macua I, Torres G, Casado FJ, Pastor-Anglada M. Link between high-affinity adenosine concentrative nucleoside transporter-2 (CNT2) and energy metabolism in intestinal and liver parenchymal cells. J Cell Physiol 2010; 225:620-30. [PMID: 20506327 DOI: 10.1002/jcp.22254] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Concentrative nucleoside transporter 2 (CNT2) is a high-affinity adenosine transporter that may play physiological roles beyond nucleoside salvage. Previous reports relate CNT2 function to modulation of purinergic signaling and energy metabolism in intestinal and liver parenchymal cells (Duflot et al., 2004, Mol Cell Biol 24:2710-2719; Aymerich et al., 2006, J Cell Sci 119:1612-1621). In the present study, to further examine the link between CNT2 and energy metabolism, CNT2 protein partners were identified using the bacterial two-hybrid and GST pull-down approaches. The N-terminal segment of CNT2 was used as bait, since proteins lacking this domain display impaired plasma membrane insertion and intracellular retention. Glucose-regulated protein 58 (GRP58) was identified as a potential rCNT2 partner in pull-down experiments. Two-hybrid screening performed against a liver human cDNA library led to the identification of aldolase B as another hCNT2 partner. Aldolase B-RFP and endogenous GRP58 separately co-localized with CNT2 in HeLa cells transfected with YFPrCNT2. CNT2 interaction with GRP58 was validated using co-immunoprecipitation experiments. In HeLa cells, fluorescence resonance energy transfer (FRET) efficiency increased upon fructose addition, consistent with a transient interaction between aldolase B and the transporter. The physiological basis for in vivo interactions was derived from experiments in which GRP58 was inhibited or overexpressed and aldolase B activity stimulated towards glycolysis. GRP58 appeared to be a negative effector of CNT2 function, whereas aldolase B flux modulated CNT2 activity via a mechanism involving acquisition of higher affinity for its substrates. These findings support the theory that CNT2 plays roles other than salvage and establishes links with energy metabolism.
Collapse
Affiliation(s)
- Isabel Huber-Ruano
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona and CIBER EHD, Barcelona, Spain
| | | | | | | | | |
Collapse
|
24
|
Plotnik DA, Emerick LE, Krohn KA, Unadkat JD, Schwartz JL. Different modes of transport for 3H-thymidine, 3H-FLT, and 3H-FMAU in proliferating and nonproliferating human tumor cells. J Nucl Med 2010; 51:1464-71. [PMID: 20720049 DOI: 10.2967/jnumed.110.076794] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The basis for the use of nucleoside tracers in PET is that activity of the cell-growth-dependent enzyme thymidine kinase 1 is the rate-limiting factor driving tracer retention in tumors. Recent publications suggest that nucleoside transporters might influence uptake and thereby affect the tracer signal in vivo. Understanding transport mechanisms for different nucleoside PET tracers is important for evaluating clinical results. This study examined the relative role of different nucleoside transport mechanisms in uptake and retention of [methyl-(3)H]-3'-deoxy-3'-fluorothymidine ((3)H-FLT), [methyl-(3)H]-thymidine ((3)H-thymidine), and (3)H-1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)-5-methyluracil ((3)H-FMAU). METHODS Transport of (3)H-FLT, (3)H-thymidine, and (3)H-FMAU was examined in a single human adenocarcinoma cell line, A549, under both nongrowth and exponential-growth conditions. RESULTS (3)H-Thymidine transport was dominated by human equilibrative nucleoside transporter 1 (hENT1) under both growth conditions. (3)H-FLT was also transported by hENT1, but passive diffusion dominated its transport. (3)H-FMAU transport was dominated by human equilibrative nucleoside transporter 2. Cell membrane levels of hENT1 increased in cells under exponential growth, and this increase was associated with a more rapid rate of uptake for both (3)H-thymidine and (3)H-FLT. (3)H-FMAU transport was not affected by changes in growth conditions. All 3 tracers concentrated in the plateau phase, nonproliferating cells at levels many-fold greater than their concentration in buffer, in part because of low levels of nucleoside metabolism, which inhibited tracer efflux. CONCLUSION Transport mechanisms are not the same for (3)H-thymidine, (3)H-FLT, and (3)H-FMAU. Levels of hENT1, an important transporter of (3)H-FLT and (3)H-thymidine, increase as proliferating cells enter the cell cycle.
Collapse
Affiliation(s)
- David A Plotnik
- Department of Radiation Oncology, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | |
Collapse
|
25
|
Errasti-Murugarren E, Molina-Arcas M, Casado FJ, Pastor-Anglada M. The human concentrative nucleoside transporter-3 C602R variant shows impaired sorting to lipid rafts and altered specificity for nucleoside-derived drugs. Mol Pharmacol 2010; 78:157-65. [PMID: 20421346 DOI: 10.1124/mol.110.063552] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The human concentrative nucleoside transporter-3 C602R (hCNT3C602R), a recently identified human concentrative nucleoside transporter-3 (hCNT3) variant, has been shown to interact with natural nucleosides with apparent K(m) values similar to those of the wild-type transporter, although binding of one of the two sodium ions required for nucleoside translocation is impaired, resulting in decreased V(max) values (Mol Pharmacol 73:379-386, 2008). We have further analyzed the properties of this hCNT3 variant by determining its localization in plasma membrane lipid domains and its interaction with nucleoside-derived drugs used in anticancer and antiviral therapies. When expressed heterologously in HeLa cells, wild-type hCNT3 localized to both lipid raft and nonlipid raft domains. Treatment of cells with the cholesterol-depleting agent methyl-beta-cyclodextrin resulted in a marked decrease in hCNT3-related transport activity that was associated with the loss of wild-type hCNT3 from lipid rafts. It is noteworthy that although exogenously expressed hCNT3C602R was present in nonlipid raft domains at a level similar to that of the wild-type transporter, the mutant transporter was present at much lower amounts in lipid rafts. A substrate profile analysis showed that interactions with a variety of nucleoside-derived drugs were altered in the hCNT3C602R variant and revealed that sugar hydroxyl residues are key structural determinants for substrate recognition by the hCNT3C602R variant.
Collapse
Affiliation(s)
- Ekaitz Errasti-Murugarren
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona and CIBER EHD, Avda Diagonal 645, Edifici annex, Planta-1, 08028 Barcelona, Spain
| | | | | | | |
Collapse
|
26
|
Striking species difference in the contribution of concentrative nucleoside transporter 2 to nucleoside uptake between mouse and rat hepatocytes. Antimicrob Agents Chemother 2010; 54:3035-8. [PMID: 20421393 DOI: 10.1128/aac.00010-10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Concentrative nucleoside transporter 2 (CNT2) (encoded by the SLC28A2 gene) transports various antiviral or antitumor purine nucleoside analogs to be involved in their pharmacokinetics and pharmacological actions. The results of our study showed that mouse hepatocytes hardly expressed CNT2 mRNA and no CNT2-dependent nucleoside uptake was observed, while rat hepatocytes exhibited high CNT2-dependent nucleoside uptake activity levels with abundant CNT2 mRNA expression. We concluded that CNT2 contributes considerably to nucleoside uptake in rat hepatocytes but not in mouse hepatocytes.
Collapse
|
27
|
Govindarajan R, Endres CJ, Whittington D, LeCluyse E, Pastor-Anglada M, Tse CM, Unadkat JD. Expression and hepatobiliary transport characteristics of the concentrative and equilibrative nucleoside transporters in sandwich-cultured human hepatocytes. Am J Physiol Gastrointest Liver Physiol 2008; 295:G570-80. [PMID: 18635603 PMCID: PMC2536788 DOI: 10.1152/ajpgi.00542.2007] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We previously reported that both the concentrative (hCNT) and equilibrative (hENT) nucleoside transporters are expressed in the human liver (21). Here we report a study that investigated the expression of these transporters (transcripts and proteins) and their role in the hepatobiliary transport of nucleosides/nucleoside drugs using sandwich-cultured human hepatocytes. In the hepatic tissue, the rank order of the mRNA expression of the transporters was hCNT1 approximately hENT1>hENT2 approximately hCNT2>hCNT3. In sandwich-cultured hepatocytes, the mRNA expression of hCNT2 and hENT2 was comparable to that in hepatic tissue, whereas the expression of corresponding transporters in the two-dimensional hepatocyte cultures was lower. Colocalization studies demonstrated predominant localization of these transporters at the sinusoidal membrane and of hENT1, hCNT1, and hCNT2 at the canalicular membrane. In the sandwich-cultured hepatocytes, ENTs were the major contributors to the transport of thymidine (hENT1, 63%; hENT2, 23%) or guanosine (hENT1, 53%; hENT2, 24%) into the hepatocytes followed by hCNT1 (10%) for thymidine or hCNT2 (23%) for guanosine. Although ribavirin was predominately transported (89%) into the hepatocytes by hENT1, fialuridine (FIAU) was transported by both hENT1 (30%) and hCNTs (61%). The extensively metabolized natural nucleosides were not effluxed into the bile, whereas significant biliary-efflux was observed of FIAU (19%), ribavirin (30%), and formycin B (35%). We conclude that the hepatic activity of hENT1 and hCNT1/2 transporters will determine the in vivo hepatic distribution and therefore the efficacy and/or toxicity of nucleoside drugs used to treat hepatic diseases.
Collapse
Affiliation(s)
- Rajgopal Govindarajan
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher J. Endres
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dale Whittington
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward LeCluyse
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marçal Pastor-Anglada
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chung-Ming Tse
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jashvant D. Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
28
|
Pérez-Torras S, García-Manteiga J, Mercadé E, Casado FJ, Carbó N, Pastor-Anglada M, Mazo A. Adenoviral-mediated overexpression of human equilibrative nucleoside transporter 1 (hENT1) enhances gemcitabine response in human pancreatic cancer. Biochem Pharmacol 2008; 76:322-9. [PMID: 18589402 DOI: 10.1016/j.bcp.2008.05.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 05/09/2008] [Accepted: 05/13/2008] [Indexed: 01/16/2023]
Abstract
Nucleoside-derived anticancer agents must be transported across the plasma membrane as a preliminary step to their conversion into active drugs. Hence, modulation of a specific nucleoside transporter may affect bioavailability and contribute significantly to sensitizing tumor cells to these anticancer agents. We have generated and functionally characterized a new recombinant adenovirus (Ad-hENT1) that has allowed us to overexpress the equilibrative nucleoside transporter hENT1 and to analyze its effects in human pancreatic tumor cells. Overexpression of hENT1 is associated with changes in cell cycle profile, in a variable manner depending on the particular cell type, thus suggesting a metabolic link between hENT1-mediated transport processes and the enzymatic machinery responsible for intracellular nucleoside metabolism. When assayed in vivo in a human pancreatic adenocarcinoma xenograft, intratumoral Ad-hENT1 injection improved the therapeutic response to gemcitabine. In summary, hENT1 overexpression is associated with alterations in nucleoside enzymatic machinery and cell cycle progression in cultured cells and enhances gemcitabine action in vivo.
Collapse
Affiliation(s)
- Sandra Pérez-Torras
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
29
|
Errasti-Murugarren E, Cano-Soldado P, Pastor-Anglada M, Casado FJ. Functional characterization of a nucleoside-derived drug transporter variant (hCNT3C602R) showing altered sodium-binding capacity. Mol Pharmacol 2008; 73:379-86. [PMID: 17993510 DOI: 10.1124/mol.107.041848] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
A novel cloned polymorphism of the human concentrative nucleoside transporter hCNT3 was described and functionally characterized. This variant consists of a T/C transition leading to the substitution of cysteine 602 by an arginine residue in the core of transmembrane domain 13. The resulting hCNT3(C602R) protein has the same selectivity and affinity for natural nucleosides and nucleoside-derived drugs as hCNT3 but much lower concentrative capacity. The insertion of the transporter into a polarized membrane seems unaffected in the variant. In a preliminary survey of a typical Spanish population, this variant showed an allelic frequency of 1%. The functional impairment of the hCNT3(C602R) polymorphism is attributable to the presence of an arginine rather than the loss of a cysteine at position 602, because an engineered hCNT3 protein with a serine residue at this position (hCNT3(C602S)) and hCNT3 have similar kinetic parameters. The sodium activation kinetic analysis of both transporters revealed a variation in the affinity for sodium and a shift in the Hill coefficient that could be consistent with a stoichiometry of 2:1 and 1:1 sodium/nucleoside, for hCNT3 and hCNT3(C602R), respectively. In conclusion, the presence of an arginine residue in the core of transmembrane domain 13 is responsible for the different sodium affinity showed by the polymorphic transporter compared with the reference transporter. Individuals with the hCNT3(C602R) variant might show a lower nucleoside and nucleoside analog concentrative capacity, which could be clinically relevant.
Collapse
Affiliation(s)
- Ekaitz Errasti-Murugarren
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal, 645 E-08028 Barcelona, Spain
| | | | | | | |
Collapse
|
30
|
Rose JB, Coe IR. Physiology of Nucleoside Transporters: Back to the Future. . . . Physiology (Bethesda) 2008; 23:41-8. [DOI: 10.1152/physiol.00036.2007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nucleoside transporters (NTs) are integral membrane proteins responsible for mediating and facilitating the flux of nucleosides and nucleobases across cellular membranes. NTs are also responsible for the uptake of nucleoside analog drugs used in the treatment of cancer and viral infections, and they are the target of certain compounds used in the treatment of some types of cardiovascular disease. The important role of NTs as drug transporters and therapeutic targets has necessarily led to intense interest into their structure and function and the relationship between these proteins and drug efficacy. In contrast, we still know relatively little about the fundamental physiology of NTs. In this review, we discuss various aspects of the physiology of NTs in mammalian systems, particularly noting tissues and cells where there has been little recent research. Our central thesis is reference back to some of the older literature, combined with current findings, will provide direction for future research into NT physiology that will lead to a fuller understanding of the role of these intriguing proteins in the everyday lives of cells, tissues, organs, and whole animals.
Collapse
Affiliation(s)
- Jennifer B. Rose
- Department of Biology, York University, Toronto, Ontario, Canada,
| | - Imogen R. Coe
- Department of Biology, York University, Toronto, Ontario, Canada,
| |
Collapse
|
31
|
Govindarajan R, Bakken AH, Hudkins KL, Lai Y, Casado FJ, Pastor-Anglada M, Tse CM, Hayashi J, Unadkat JD. In situ hybridization and immunolocalization of concentrative and equilibrative nucleoside transporters in the human intestine, liver, kidneys, and placenta. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1809-22. [PMID: 17761511 DOI: 10.1152/ajpregu.00293.2007] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To better understand the role of human equilibrative (hENTs) and concentrative (hCNTs) nucleoside transporters in physiology and pharmacology, we investigated the regional, cellular, and spatial distribution of two hCNTs (hCNT1 and hCNT2) and two hENTs (hENT1 and hENT2) in four human tissues. Using in situ hybridization and immunohistochemical techniques, we found that the duodenum expressed hCNT1 and hCNT2 mRNAs in enterocytes and hENT1 and hENT2 mRNAs in crypt cells. In these cells, the hCNT and hENT proteins were predominantly localized in the apical and lateral membrane, respectively. Hepatocytes expressed higher levels of mRNAs of hENT1, hCNT1, and hENT2 than of hCNT2 and expressed all these proteins at hepatocyte cell borders and in the cytoplasm. While the kidney expressed hCNT1 and hCNT2 mRNAs in the proximal tubules, hENT1 and hENT2 mRNAs were present in the distal tubules, glomeruli, endothelial cells, and vascular smooth muscle cells. Proximal tubules adjacent to corticomedullary junctions expressed hENT1, hCNT1, and hCNT2 mRNA. Immunolocalization studies revealed predominant localization of hCNTs in the brush-border membrane of the proximal tubular epithelial cells and hENTs in the basolateral membrane of the distal tubular epithelial cells. Chorionic villi sections of human term placenta expressed mRNAs and proteins for hENT1 and hENT2 but only mRNA for hCNT2. Immunolocalization studies showed presence of hENT1 in the brush-border membrane of the syncytiotrophoblasts. These data are critical for a better understanding of the role of nucleoside transporters in the physiological and pharmacological effects of nucleosides and nucleoside drugs, respectively.
Collapse
Affiliation(s)
- Rajgopal Govindarajan
- Dept. of Pharmaceutics, University of Washington, Box 357610, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Errasti-Murugarren E, Pastor-Anglada M, Casado FJ. Role of CNT3 in the transepithelial flux of nucleosides and nucleoside-derived drugs. J Physiol 2007; 582:1249-60. [PMID: 17412768 PMCID: PMC2075247 DOI: 10.1113/jphysiol.2007.130138] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
We examined the role of the concentrative nucleoside transporter CNT3 in the establishment of a transepithelial flux of natural nucleosides and their pharmacologically active derivatives in renal epithelial cell lines. Murine PCT cells grown on a transwell dish showed endogenous CNT3 activity at their apical membrane that was responsible for the sodium-dependent transepithelial flux of both purine and pyrimidine nucleosides. hCNT3 was also identified in human kidney and its role in the transport of nucleosides was tested. To this end, MDCK cells, lacking endogenous CNT3 activity, were genetically engineered to express the human orthologue of CNT3 (hCNT3-MDCK cells). In these cells, hCNT3 was inserted into the apical membrane, thus generating, as for PCT cells, a transepithelial flux of both nucleosides and nucleoside-derived drugs. Apical-to-basolateral transepithelial flux was present in all cells expressing a functional CNT3 transporter and was significantly higher than that found either in PCT cells in absence of sodium or in mock-transfected MDCK cells. Nevertheless in all cases a significant amount of the transported nucleoside was retained and transformed inside cells. However release to the opposite compartment was CNT3 dependent, not only in terms of absolute flux (much higher when an apical CNT3 transporter was active) but also regarding metabolic transformations of the apically absorbed nucleosides. These results underline a critical role of CNT3 in the renal reabsorption of nucleosides and their derivatives as well as in their intracellular metabolism.
Collapse
Affiliation(s)
- Ekaitz Errasti-Murugarren
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 645, Edifici annex, Planta-1, E-08028 Barcelona, Spain
| | | | | |
Collapse
|
33
|
Löffler M, Morote-Garcia JC, Eltzschig SA, Coe IR, Eltzschig HK. Physiological roles of vascular nucleoside transporters. Arterioscler Thromb Vasc Biol 2007; 27:1004-13. [PMID: 17332491 DOI: 10.1161/atvbaha.106.126714] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Nucleoside transporters (NTs) comprise 2 widely expressed families, the equilibrative nucleoside transporters (diffusion-limited channels) and concentrative nucleoside transporters (sodium-dependent transporters). Because of their anatomic position at the blood-tissue interface, vascular NTs are in an ideal position to influence vascular nucleoside levels, particularly adenosine, which among others plays an important role in tissue protection during acute injury. For example, endothelial NTs contribute to preserving the vascular integrity during conditions of limited oxygen availability (hypoxia). Indeed, hypoxia-inducible factor-1-dependent repression of NTs results in enhanced extracellular adenosine signaling and thus attenuates hypoxia-associated increases in vascular leakage. In addition, vascular NTs also contribute to cardiac ischemic preconditioning, coronary vasodilation, and inhibition of platelet aggregation. Moreover, vascular nucleoside uptake via NTs is important for nucleoside recovery, particularly in cells lacking de novo nucleotide synthesis pathways (erythrocytes, leukocytes). Taken together, vascular NTs are critical in modulating adenosine-mediated responses during conditions such as inflammation or hypoxia.
Collapse
Affiliation(s)
- Michaela Löffler
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Waldhörnle Str. 22, 72072, Tübingen, Germany
| | | | | | | | | |
Collapse
|
34
|
Pastor-Anglada M, Errasti-Murugarren E, Aymerich I, Casado FJ. Concentrative nucleoside transporters (CNTs) in epithelia: from absorption to cell signaling. J Physiol Biochem 2007; 63:97-110. [PMID: 17722647 DOI: 10.1007/bf03174089] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Concentrative and Equilibrative Nucleoside Transporter proteins (CNT and ENT, respectively) are encoded by gene families SLC28 and SLC29. They mediate the uptake of natural nucleosides and a variety of nucleoside-derived drugs, mostly used in anticancer therapy. CNT and ENT proteins are mostly localized in the apical and basolateral sides, respectively, in (re)absorptive epithelia. This anatomic distribution determines nucleoside and nucleoside-derived vectorial flux. CNT expression (particularly CNT2) is associated with differentiation and is also nutritionally regulated in intestinal epithelia, whereas ENT protein amounts (mostly ENT1) are increased when cells are exposed to proliferative stimuli such as EGF, TGF-alpha or wounding. Although all these features suggest a role for NT proteins in nucleoside salvage and (re)absorption, recent data demonstrate that CNT2 might be under purinergic control, in a manner that is dependent on energy metabolism. A physiological link between CNT2 function and intracellular metabolism is also supported by the evidence that extracellular adenosine can activate the AMP-dependent kinase (AMPK), by a mechanism which relies upon adenosine transport and phosphorylation. Thus the complex pattern of NT isoform expression in mammalian cells can fulfill physiological roles other than salvage.
Collapse
Affiliation(s)
- M Pastor-Anglada
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona, Spain.
| | | | | | | |
Collapse
|
35
|
Fernández-Veledo S, Jover R, Casado FJ, Gómez-Lechón MJ, Pastor-Anglada M. Transcription factors involved in the expression of SLC28 genes in human liver parenchymal cells. Biochem Biophys Res Commun 2007; 353:381-8. [PMID: 17187757 DOI: 10.1016/j.bbrc.2006.12.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2006] [Accepted: 12/05/2006] [Indexed: 11/30/2022]
Abstract
Human nucleoside transporters are encoded by SLC28 (hCNTs) and SLC29 (hENTs) genes. These proteins mediate the uptake of anticancer and some antiviral drugs and are also suitable candidates to facilitate nucleoside-derived drug uptake into hepatocytes for detoxification. Despite the putative relevance of these genes in liver physiology, the human SLC28 and SLC29 expression pattern is not known and suitable cell models are not available. These issues have been addressed by examining NT expression in human liver and primary cultures of human hepatocytes. Moreover, the effect of specific liver enriched transcription factors (LETFs) in hCNTs expression has been analyzed. Human hepatocytes express hCNT1, hCNT2, hENT1, and hENT2. Loss of the hepatic phenotype in primary culture is associated with a decrease in hCNT1 and hCNT2 mRNA levels. Selected LETFs are involved in the regulation of SLC28 genes in an isoform-specific manner. HNF4alpha is a major determinant of SLC28A1 expression, whereas C/EBPalpha and HNF3gamma modulate SLC28A2 gene expression.
Collapse
Affiliation(s)
- Sonia Fernández-Veledo
- Departament de Bioquímica i Biologia Molecular, Facultat de Biología, Universitat de Barcelona, and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Diagonal 645, E-08028 Barcelona, Spain
| | | | | | | | | |
Collapse
|
36
|
Potassium channels: new targets in cancer therapy. ACTA ACUST UNITED AC 2006; 30:375-85. [PMID: 16971052 DOI: 10.1016/j.cdp.2006.06.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2006] [Indexed: 01/01/2023]
Abstract
BACKGROUND Potassium channels (KCh) are the most diverse and ubiquitous class of ion channels. KCh control membrane potential and contribute to nerve and cardiac action potentials and neurotransmitter release. KCh are also involved in insulin release, differentiation, activation, proliferation, apoptosis, and several other physiological functions. The aim of this review is to provide an updated overview of the KCh role during the cell growth. Their potential use as pharmacological targets in cancer therapies is also discussed. METHODS We searched PubMed (up to 2005) and identified relevant articles. Reprints were mainly obtained by on line subscription. Additional sources were identified through cross-referencing and obtained from Library services. RESULTS KCh are responsible for some neurological and cardiovascular diseases and for a new medical discipline, channelopathies. Their role in congenital deafness, multiple sclerosis, episodic ataxia, LQT syndrome and diabetes has been proven. Furthermore, a large body of information suggests that KCh play a role in the cell cycle progression, and it is now accepted that cells require KCh to proliferate. Thus, KCh expression has been studied in a number of tumours and cancer cells. CONCLUSIONS Cancer is far from being considered a channelopathy. However, it seems appropriate to take into account the involvement of KCh in cancer progression and pathology when developing new strategies for cancer therapy.
Collapse
|
37
|
Gloeckner-Hofmann K, Guillén-Gómez E, Schmidtgen C, Porstmann R, Ziegler R, Stoss O, Casado FJ, Rüschoff J, Pastor-Anglada M. Expression of the High-Affinity Fluoropyrimidine-Preferring Nucleoside Transporter hCNT1 Correlates with Decreased Disease-Free Survival in Breast Cancer. Oncology 2006; 70:238-44. [PMID: 16837820 DOI: 10.1159/000094541] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2005] [Accepted: 05/18/2006] [Indexed: 01/14/2023]
Abstract
PURPOSE Nucleoside and nucleobase derivatives are currently used in the treatment of a variety of solid tumors; however, the role of plasma membrane transporters as biomarkers of drug metabolism has not been fully addressed. Thus, the purpose of this study was to determine whether the concentrative nucleoside transporter hCNT1 is a predictive marker of therapeutic response. METHODS We studied a cohort of 90 breast cancer patients who were treated with cyclophosphamide-methotrexate-5-fluorouracil after surgery and then monitored for up to 108 months. hCNT1 and enzymes associated with nucleotide metabolism (thymidine phosphorylase, dihydropyrimidine dehydrogenase and thymidylate synthase) were assessed immunohistochemically in tissue samples. RESULTS Human CNT1 presence was mostly cytoplasmic, with some nuclear staining. The percentage of hCNT1-positive cells correlated positively with the expression of thymidine phosphorylase and dihydropyrimidine dehydrogenase. Nuclear staining correlated negatively with decreased disease-free survival, whereas the percentage of hCNT1-positive cells correlated positively with reduced long-term survival, with the hCNT1-positive index (>80%) being indicative of poor prognosis. A relative risk of relapse was associated with high hCNT1-positive indexes, whereas when this parameter was combined with the nodal status (positive), a high risk of relapse was found, suggesting that both parameters may reflect a poor prognosis. CONCLUSIONS These results indicate that the expression of the high-affinity concentrative nucleoside transporter hCNT1 has a prognostic value in determining disease-free survival and risk of relapse in breast cancer patients undergoing surgery followed by cyclophosphamide-methotrexate-5-fluorouracil chemotherapy.
Collapse
|
38
|
Tozzi MG, Camici M, Mascia L, Sgarrella F, Ipata PL. Pentose phosphates in nucleoside interconversion and catabolism. FEBS J 2006; 273:1089-101. [PMID: 16519676 DOI: 10.1111/j.1742-4658.2006.05155.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ribose phosphates are either synthesized through the oxidative branch of the pentose phosphate pathway, or are supplied by nucleoside phosphorylases. The two main pentose phosphates, ribose-5-phosphate and ribose-1-phosphate, are readily interconverted by the action of phosphopentomutase. Ribose-5-phosphate is the direct precursor of 5-phosphoribosyl-1-pyrophosphate, for both de novo and 'salvage' synthesis of nucleotides. Phosphorolysis of deoxyribonucleosides is the main source of deoxyribose phosphates, which are interconvertible, through the action of phosphopentomutase. The pentose moiety of all nucleosides can serve as a carbon and energy source. During the past decade, extensive advances have been made in elucidating the pathways by which the pentose phosphates, arising from nucleoside phosphorolysis, are either recycled, without opening of their furanosidic ring, or catabolized as a carbon and energy source. We review herein the experimental knowledge on the molecular mechanisms by which (a) ribose-1-phosphate, produced by purine nucleoside phosphorylase acting catabolically, is either anabolized for pyrimidine salvage and 5-fluorouracil activation, with uridine phosphorylase acting anabolically, or recycled for nucleoside and base interconversion; (b) the nucleosides can be regarded, both in bacteria and in eukaryotic cells, as carriers of sugars, that are made available though the action of nucleoside phosphorylases. In bacteria, catabolism of nucleosides, when suitable carbon and energy sources are not available, is accomplished by a battery of nucleoside transporters and of inducible catabolic enzymes for purine and pyrimidine nucleosides and for pentose phosphates. In eukaryotic cells, the modulation of pentose phosphate production by nucleoside catabolism seems to be affected by developmental and physiological factors on enzyme levels.
Collapse
Affiliation(s)
- Maria G Tozzi
- Dipartimento di Biologia, Laboratorio di Biochimica, Pisa, Italy
| | | | | | | | | |
Collapse
|
39
|
Fernández-Veledo S, Huber-Ruano I, Aymerich I, Duflot S, Casado F, Pastor-Anglada M. Bile acids alter the subcellular localization of CNT2 (concentrative nucleoside cotransporter) and increase CNT2-related transport activity in liver parenchymal cells. Biochem J 2006; 395:337-44. [PMID: 16390326 PMCID: PMC1422760 DOI: 10.1042/bj20051232] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CNT2 (concentrative nucleoside cotransporter) is a plasma membrane high-affinity Na+-coupled adenosine transporter, also localized in intracellular structures. This transporter protein may play additional roles other than nucleoside salvage, since it has recently been shown to be under purinergic control via K(ATP) channels, by a mechanism that does not seem to involve changes in its subcellular localization. In an attempt to identify the agents that promote CNT2 trafficking, bile acids were found to increase CNT2-related transport activity in a K(ATP) channel-independent manner in both Fao hepatoma and rat liver parenchymal cells. A maximum effect was recorded after treatment with hydrophilic anions such as TCA (taurocholate). However, this effect did not involve changes in the amount of CNT2 protein, it was instead associated with a subcellular redistribution of CNT2, resulting in an accumulation of the transporter at the plasma membrane. This was deduced from subcellular fractionation studies, biotinylation of plasma membrane proteins and subsequent CNT2 detection in streptavidin precipitates and in vivo confocal microscopic analysis of the distribution of a YFP (yellow fluorescent protein)-CNT2 construct. The induction of CNT2 translocation, triggered by TCA, was inhibited by wortmannin, dibutyryl-AMPc, PD98059 and colchicine, thus suggesting the involvement of the PI3K/ERK (phosphoinositide 3-kinase/extracellular-signal related kinase) pathway in microtubule-dependent activation of recombinant CNT2. These are novel effects of bile-acid physiology and provide the first evidence for short-term regulation of CNT2 translocation into and from the plasma membrane.
Collapse
Affiliation(s)
- Sonia Fernández-Veledo
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Diagonal 645, E-08028 Barcelona, Spain
| | - Isabel Huber-Ruano
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Diagonal 645, E-08028 Barcelona, Spain
| | - Ivette Aymerich
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Diagonal 645, E-08028 Barcelona, Spain
| | - Sylvie Duflot
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Diagonal 645, E-08028 Barcelona, Spain
| | - F. Javier Casado
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Diagonal 645, E-08028 Barcelona, Spain
| | - Marçal Pastor-Anglada
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Diagonal 645, E-08028 Barcelona, Spain
- To whom correspondence should be addressed (email )
| |
Collapse
|
40
|
Aymerich I, Foufelle F, Ferré P, Casado FJ, Pastor-Anglada M. Extracellular adenosine activates AMP-dependent protein kinase (AMPK). J Cell Sci 2006; 119:1612-21. [PMID: 16569664 DOI: 10.1242/jcs.02865] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a heterotrimeric complex that senses intracellular energy status and exerts rapid regulation on energy-demanding and -consuming metabolic pathways. Although alterations in the intracellular adenosine nucleotide pool are traditionally assumed to be the consequence of changes in energy metabolism, in this study we have addressed the question of whether extracellular adenosine contributes to AMPK regulation. In the intestinal rat epithelial cell line IEC-6, addition of adenosine rapidly increases AMP intracellular concentrations and upregulates alpha1AMPK, thus promoting phosphorylation of its downstream target acetyl-CoA carboxylase (ACC). The effect of adenosine on AMPK signaling is completely blocked by transducing IEC-6 cells with an adenoviral vector expressing a mutated alpha1 subunit, resulting in a dominant-negative effect on endogenous AMPK activity. These effects are blocked by 5'-iodotubercidine (5'-ITU), an inhibitor of adenosine kinase. Moreover, inhibition of adenosine transport through the concentrative adenosine plasma membrane transporter CNT2 with formycin B results in the blockade of adenosine-mediated AMPK signaling. Extracellular adenosine is equally able to activate AMPK and promote ACC phosphorylation in liver parenchymal cell models in a manner that is also inhibited by 5'-ITU. In summary, this study shows that adenosine, when added at physiological concentrations, activates AMPK and promotes ACC phosphorylation. Adenosine must be transported and phosphorylated to exert its action. Thus, nucleoside transporters might be novel players in the complex regulation of AMPK and energy metabolism.
Collapse
Affiliation(s)
- Ivette Aymerich
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, 08071 Barcelona, Spain
| | | | | | | | | |
Collapse
|
41
|
Rodríguez-Mulero S, Errasti-Murugarren E, Ballarín J, Felipe A, Doucet A, Casado FJ, Pastor-Anglada M. Expression of concentrative nucleoside transporters SLC28 (CNT1, CNT2, and CNT3) along the rat nephron: Effect of diabetes. Kidney Int 2005; 68:665-72. [PMID: 16014043 DOI: 10.1111/j.1523-1755.2005.00444.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The renal reabsorption of natural nucleosides and a variety of nucleoside-derived drugs relies on the function of the apically located, Na(+)-dependent, concentrative nucleoside transporters CNT1, CNT2, and CNT3 (SLC28A1, SLC28A2, and SLC28A3). The aims of this study were to determine the segmental localization of the three SLC28 family members and to establish whether streptozotocin-induced diabetes alters their expression. METHODS SLC28 expression was measured by real-time polymerase chain reaction (PCR) on microdissected sections of rat nephrons. Diabetes was induced by streptozotocin treatment and the biochemical profiles of control, diabetic, and insulin-treated rats were established. The effect of diabetes on SLC28 expression was assessed in those segments that significantly express SLC28 genes. RESULTS CNT1-3 mRNAs were expressed in the proximal tubule and glomerulus. In addition, CNT2 and CNT3 mRNAs were expressed in the outer medullary and cortical collecting duct, respectively. Diabetes reduced expression of the three CNTs in almost all nephron segments, and this effect was not prevented by an insulin treatment that normalized all blood and urine parameters. Diabetes increased CNT1 and CNT3 expression in the glomerulus and insulin treatment decreased it. CONCLUSION The relative distribution of SLC28 gene expression suggests a role for the proximal tubule in renal nucleoside clearance and an accessory role for CNT2 and CNT3, in adenosine-mediated regulation of collecting duct functions. Diabetes probably may impair nucleoside clearance independently of insulin.
Collapse
|
42
|
Lum JJ, DeBerardinis RJ, Thompson CB. Autophagy in metazoans: cell survival in the land of plenty. Nat Rev Mol Cell Biol 2005; 6:439-48. [PMID: 15928708 DOI: 10.1038/nrm1660] [Citation(s) in RCA: 605] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cells require a constant supply of macromolecular precursors and oxidizable substrates to maintain viability. Unicellular eukaryotes lack the ability to regulate nutrient concentrations in their extracellular environment. So when environmental nutrients are depleted, these organisms catabolize existing cytoplasmic components to support ATP production to maintain survival, a process known as autophagy. By contrast, the environment of metazoans normally contains abundant extracellular nutrients, but a cell's ability to take up these nutrients is controlled by growth factor signal transduction. Despite evolving the ability to maintain a constant supply of extracellular nutrients, metazoans have retained a complete set of autophagy genes. The physiological relevance of autophagy in such species is just beginning to be explored.
Collapse
Affiliation(s)
- Julian J Lum
- Abramson Family Cancer Research Institute, Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Philadelphia 19104, USA
| | | | | |
Collapse
|
43
|
Aymerich I, Pastor-Anglada M, Casado FJ. Long term endocrine regulation of nucleoside transporters in rat intestinal epithelial cells. ACTA ACUST UNITED AC 2005; 124:505-12. [PMID: 15504900 PMCID: PMC2234001 DOI: 10.1085/jgp.200409086] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We studied the regulation of nucleoside transporters in intestinal epithelial cells upon exposure to either differentiating or proliferative agents. Rat intestinal epithelial cells (line IEC-6) were incubated in the presence of differentiating (glucocorticoids) or proliferative (EGF and TGF-α) agents. Nucleoside uptake rates and nucleoside transporter protein and mRNA levels were assessed. The signal transduction pathways used by the proliferative stimuli were analyzed. We found that glucocorticoids induce an increase in sodium-dependent, concentrative nucleoside transport rates and in protein and mRNA levels of both rCNT2 and rCNT1, with negligible effects on the equilibrative transporters. EGF and TGF-α induce an increase in the equilibrative transport rate, mostly accounted for by an increase in rENT1 activity and mRNA levels, rENT2 mRNA levels remaining unaltered. This effect is mimicked by another proliferative stimulus that functions as an in vitro model of epithelial wounding. Here, rENT1 activity and mRNA levels are also increased, although the signal transduction pathways used by the two stimuli are different. We concluded that differentiation of rat intestinal epithelial cells is accompanied by increased mature enterocyte features, such as concentrative nucleoside transport (located at the brush border membrane of the enterocyte), thus preparing the cell for its ultimate absorptive function. A proliferative stimulus induces the equilibrative nucleoside activities (mostly through ENT1) known to be located at the basolateral membrane, allowing the uptake of nucleosides from the bloodstream for the increased demands of the proliferating cell.
Collapse
Affiliation(s)
- Ivette Aymerich
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal, 645, 08071 Barcelona, Spain
| | | | | |
Collapse
|
44
|
Pastor-Anglada M, Cano-Soldado P, Molina-Arcas M, Lostao MP, Larráyoz I, Martínez-Picado J, Casado FJ. Cell entry and export of nucleoside analogues. Virus Res 2005; 107:151-64. [PMID: 15649561 DOI: 10.1016/j.virusres.2004.11.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Some nucleoside analogues currently used as antiretroviral agents might promote mutagenesis besides their putative ability to interfere with endogenous nucleotide metabolism and/or inhibit viral transcription. The intracellular concentration of nucleosides and nucleobases is to some extent the result of the metabolic background of the specific cell line used for infection studies, its particular suit of enzymes and transporters. This review focuses on the transporter-mediated pathways implicated in either the uptake or the efflux of nucleoside- and nucleobase-derivatives. From a biochemical point of view, four different types of transport processes for nucleoside-related antiviral drugs have been described: (1) equilibrative uniport, (2) substrate exchange, (3) concentrative Na+- or H+-dependent uptake and finally, (4) substrate export through primary ATP-dependent active efflux pumps. These mechanisms are mainly related to the following set of transporter families: Concentrative Nucleoside Transporter (CNT), Equilibrative Nucleoside Transporter (ENT), Organic Anion Transporter (OAT) and Organic Cation Transporter (OCT), Peptide Transporter (PEPT) and Multidrug Resistance Protein (MRP). The basic properties of these carrier proteins and their respective role in the transport across the plasma membrane of nucleoside-derived antiviral drugs are reviewed.
Collapse
Affiliation(s)
- Marçal Pastor-Anglada
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Diagonal 645, 08028 Barcelona, Spain.
| | | | | | | | | | | | | |
Collapse
|
45
|
Aymerich I, Duflot S, Fernández-Veledo S, Guillén-Gómez E, Huber-Ruano I, Casado FJ, Pastor-Anglada M. The concentrative nucleoside transporter family (SLC28): new roles beyond salvage? Biochem Soc Trans 2005; 33:216-9. [PMID: 15667311 DOI: 10.1042/bst0330216] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The concentrative nucleoside transporter (CNT) family (SLC28) has three members: SLC28A1 (CNT1), SLC28A2 (CNT2) and SLC28A3 (CNT3). The CNT1 and CNT2 transporters are co-expressed in liver parenchymal cells and macrophages, two suitable models in which to study cell cycle progression. Despite initial observations suggesting that these transporter proteins might contribute to nucleoside salvage during proliferation, their subcellular localization and regulatory properties suggest alternative roles in cell physiology. In particular, CNT2 is a suitable candidate for modulation of purinergic responses, since it is under the control of the adenosine 1 receptor. Increasing evidence also suggests a role for CNT2 in energy metabolism, since its activation relies on the opening of ATP-sensitive K+ channels. Animal and cell models genetically modified to alter nucleoside transporter expression levels may help to elucidate the particular roles of CNT proteins in cell physiology.
Collapse
Affiliation(s)
- I Aymerich
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Diagonal 645, 08071 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
46
|
Sakowicz M, Szutowicz A, Pawelczyk T. Insulin and glucose induced changes in expression level of nucleoside transporters and adenosine transport in rat T lymphocytes. Biochem Pharmacol 2004; 68:1309-20. [PMID: 15345320 DOI: 10.1016/j.bcp.2004.06.020] [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] [Received: 05/07/2004] [Accepted: 06/14/2004] [Indexed: 11/24/2022]
Abstract
Adenosine is an endogenous agent exerting potent action on the immune system including regulation of lymphocyte functioning. Impaired T lymphocyte functioning is a common feature of diabetes. The aims of this study were to examine the effects of glucose and insulin on nucleoside transporters (NT) expression level and adenosine (Ado) transport in rat T lymphocytes cultured under the defined concentrations of glucose and insulin. Performed experiments revealed that rat T lymphocytes expressed the equilibrative nucleoside transporter type 1 and 2 (rENT1, rENT2) and concentrative nucleoside transporter type 2 (rCNT2). The mRNA levels of rENT2 and rCNT2 were highly dependent on insulin but were not affected by changes in extracellular glucose concentration. Exposition of T cells to 10nM insulin resulted in 73% increase in rENT2 mRNA and 50% decrease in the rCNT2 mRNA level. The level of rENT1 mRNA was sensitive to extracellular glucose concentration but not to insulin. The highest differences among cells cultured in high (20mM) and low (5mM) glucose were observed in equilibrative nitrobenzylthioinosine sensitive adenosine transport, which was lowered by 65% in cells cultured at high glucose. Alterations in adenosine transport were accompanied by changes in adenosine accumulation in the cell. These results indicate that adenosine transport in rat T lymphocytes is independently and differentially regulated by glucose and insulin by means of changes in the nucleoside transporters expression level. Altered adenosine transport has a great impact on its intracellular level. This suggests that under diabetic conditions adenosine action on T lymphocytes might be altered.
Collapse
Affiliation(s)
- Monika Sakowicz
- Department of Molecular Medicine, Medical University of Gdansk, Poland
| | | | | |
Collapse
|
47
|
Fernández-Veledo S, Valdés R, Wallenius V, Casado FJ, Pastor-Anglada M. Up-regulation of the high-affinity pyrimidine-preferring nucleoside transporter concentrative nucleoside transporter 1 by tumor necrosis factor-alpha and interleukin-6 in liver parenchymal cells. J Hepatol 2004; 41:538-44. [PMID: 15464233 DOI: 10.1016/j.jhep.2004.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Revised: 05/20/2004] [Accepted: 06/09/2004] [Indexed: 01/27/2023]
Abstract
BACKGROUND/AIMS Concentrative nucleoside transporter 1 (CNT1), a high affinity transporter for pyrimidine nucleosides, is responsible for their Na+-dependent concentrative uptake into hepatocytes. Though CNT1 protein amounts increase in rat liver soon after partial hepatectomy, the physiological regulators of CNT1 expression have not yet been identified. METHODS Rat hepatoma cell lines and hepatocytes isolated from fetuses and adult rats were used to identify single agents able to up-regulate CNT1 expression and activity in liver. TNF-alpha receptor-I (TNFRI) and IL-6 knock-out mice were also used to study CNT1 regulation in vivo. RESULTS TNF-alpha and IL-6 independently induced CNT1 protein expression in cultured liver parenchymal and FAO hepatoma cells by PI-3 kinase- and ERK-dependent mechanisms, respectively. In vivo data showed that transporter protein levels were low in livers from TNFRI knock-out mice, but not in those from IL-6 deficient animals. However, IL-6 administration only partially restored CNT1 expression in the former model. CONCLUSIONS This study identifies TNF-alpha as a major in vivo modulator of the nucleoside transporter CNT1 and suggests a secondary role for IL-6 in mediating CNT1 up-regulation by TNF-alpha in vivo. Evidence is provided that two independent pathways are involved in the up-regulation of CNT1 by TNF-alpha and IL-6.
Collapse
Affiliation(s)
- Sonia Fernández-Veledo
- Department de Bioquímica i Biologia Molecular, Universitat de Barcelona, Diagonal 645, E-08028 Barcelona, Spain
| | | | | | | | | |
Collapse
|
48
|
Cano-Soldado P, Larráyoz IM, Molina-Arcas M, Casado FJ, Martinez-Picado J, Lostao MP, Pastor-Anglada M. Interaction of Nucleoside Inhibitors of HIV-1 Reverse Transcriptase with the Concentrative Nucleoside Transporter-1 (Slc28A1). Antivir Ther 2004. [DOI: 10.1177/135965350400900617] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human concentrative nucleoside transporter-1 (hCNT1) (SLC28A1) is a widely expressed, high-affinity, pyrimi-dine-preferring, nucleoside transporter implicated in the uptake of naturally occurring pyrimidine nucleosides as well as a variety of derivatives used in anticancer treatment. Its putative role in the uptake of other pyrimidine nucleoside analogues with antiviral properties has not been studied in detail to date. Here, using a hCNT1 stably transfected cell line and the two-electrode voltage-clamp technique, we have assessed the interaction of selected pyrimidine-based antiviral drugs, inhibitors of HIV-1 reverse transcriptase such as zidovudine (AZT), stavudine (d4T), lamivudine (3TC) and zalcitabine (ddC), with hCNT1. hCNT1 transports AZT and d4T with low affinity, whereas 3TC and ddC are not translocated, the latter being able to bind the transporter protein. Selectivity appears to rely mostly upon the presence of a hydroxyl group in the 3′-position of the ribose ring. Thus, hCNT1 cannot be considered a broad-selectivity pyrimidine nucleoside carrier; in fact, very slight changes in substrate structure provoke a dramatic shift in selectivity.
Collapse
Affiliation(s)
- Pedro Cano-Soldado
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Barcelona, Spain
| | - Ignacio M Larráyoz
- Departamento de Fisiología y Nutrición, Universidad de Navarra, Pamplona, Spain
| | - Míriam Molina-Arcas
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Barcelona, Spain
| | - F Javier Casado
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Barcelona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa Foundation, Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - M Pilar Lostao
- Departamento de Fisiología y Nutrición, Universidad de Navarra, Pamplona, Spain
| | - Marçal Pastor-Anglada
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
49
|
Duflot S, Riera B, Fernández-Veledo S, Casadó V, Norman RI, Casado FJ, Lluís C, Franco R, Pastor-Anglada M. ATP-sensitive K(+) channels regulate the concentrative adenosine transporter CNT2 following activation by A(1) adenosine receptors. Mol Cell Biol 2004; 24:2710-9. [PMID: 15024061 PMCID: PMC371120 DOI: 10.1128/mcb.24.7.2710-2719.2004] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This study describes a novel mechanism of regulation of the high-affinity Na(+)-dependent adenosine transporter (CNT2) via the activation of A(1) adenosine receptors (A(1)R). This regulation is mediated by the activation of ATP-sensitive K(+) (K(ATP)) channels. The high-affinity Na(+)-dependent adenosine transporter CNT2 and A(1)R are coexpressed in the basolateral domain of the rat hepatocyte plasma membrane and are colocalized in the rat hepatoma cell line FAO. The transient increase in CNT2-mediated transport activity triggered by (-)-N(6)-(2-phenylisopropyl)adenosine is fully inhibited by K(ATP) channel blockers and mimicked by a K(ATP) channel opener. A(1)R agonist activation of CNT2 occurs in both hepatocytes and FAO cells, which express Kir6.1, Kir6.2, SUR1, SUR2A, and SUR2B mRNA channel subunits. With the available antibodies against Kir6.X, SUR2A, and SUR2B, it is shown that all of these proteins colocalize with CNT2 and A(1)R in defined plasma membrane domains of FAO cells. The extent of the purinergic modulation of CNT2 is affected by the glucose concentration, a finding which indicates that glycemia and glucose metabolism may affect this cross-regulation among A(1)R, CNT2, and K(ATP) channels. These results also suggest that the activation of K(ATP) channels under metabolic stress can be mediated by the activation of A(1)R. Cell protection under these circumstances may be achieved by potentiation of the uptake of adenosine and its further metabolization to ATP. Mediation of purinergic responses and a connection between the intracellular energy status and the need for an exogenous adenosine supply are novel roles for K(ATP) channels.
Collapse
Affiliation(s)
- Sylvie Duflot
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, E-08071 Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gray JH, Owen RP, Giacomini KM. The concentrative nucleoside transporter family, SLC28. Pflugers Arch 2004; 447:728-34. [PMID: 12856181 DOI: 10.1007/s00424-003-1107-y] [Citation(s) in RCA: 306] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2003] [Revised: 05/04/2003] [Accepted: 05/04/2003] [Indexed: 10/26/2022]
Abstract
The SLC28 family consists of three subtypes of sodium-dependent, concentrative nucleoside transporters, CNT1, CNT2, and CNT3 (SLC28A1, SLC28A2, and SLC28A3, respectively), that transport both naturally occurring nucleosides and synthetic nucleoside analogs used in the treatment of various diseases. These subtypes differ in their substrate specificities: CNT1 is pyrimidine-nucleoside preferring, CNT2 is purine-nucleoside preferring, and CNT3 transports both pyrimidine and purine nucleosides. Recent studies have identified key amino acid residues that are determinants of pyrimidine and purine specificity of CNT1 and CNT2. The tissue distributions of the CNTs vary: CNT1 is localized primarily in epithelia, whereas CNT2 and CNT3 have more generalized distributions. Nucleoside transporters in the SLC28 and SLC29 families play critical roles in nucleoside salvage pathways where they mediate the first step of nucleotide biosynthesis. In addition, these transporters work in concert to terminate adenosine signaling. SLC28 family members are crucial determinants of response to a variety of anticancer and antiviral nucleoside analogs, as they modulate the entry of these analogs into target tissues. Further, this family is involved in the absorption and disposition of many nucleoside analogs. Several CNT single nucleoside polymorphisms (SNPs) have been identified, but have yet to be characterized.
Collapse
Affiliation(s)
- Jennifer H Gray
- Department of Biopharmaceutical Sciences, University of California-San Francisco, 513 Parnassus Ave., S-926, San Francisco, CA 94143-0446, USA.
| | | | | |
Collapse
|