Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Journal of Pharmacology and Experimental Therapeutics
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • Log out
  • My Cart
Journal of Pharmacology and Experimental Therapeutics

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit jpet on Facebook
  • Follow jpet on Twitter
  • Follow jpet on LinkedIn
Research ArticleMETABOLISM, TRANSPORT, AND PHARMACOGENOMICS

Expression and Functionality of Anti-Human Immunodeficiency Virus and Anticancer Drug Uptake Transporters in Immune Cells

Gerard Minuesa, Sergi Purcet, Itziar Erkizia, Míriam Molina-Arcas, Margarita Bofill, Nuria Izquierdo-Useros, F. Javier Casado, Bonaventura Clotet, Marçal Pastor-Anglada and Javier Martinez-Picado
Journal of Pharmacology and Experimental Therapeutics February 2008, 324 (2) 558-567; DOI: https://doi.org/10.1124/jpet.107.131482
Gerard Minuesa
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sergi Purcet
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Itziar Erkizia
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Míriam Molina-Arcas
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Margarita Bofill
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nuria Izquierdo-Useros
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
F. Javier Casado
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bonaventura Clotet
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marçal Pastor-Anglada
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Javier Martinez-Picado
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

Almost all drugs used in anti-human immunodeficiency virus (HIV)-1 and anticancer therapies require membrane proteins to get into the cell to develop their proper activity. Nevertheless, little is known regarding the expression and activity of specific carriers involved in the uptake of these drugs in immune cells. Here, we assessed the mRNA levels, protein expression profile, and activity of the gene families SLC28 (coding for concentrative nucleoside transporters, hCNT1–3), SLC29 (equilibrative nucleoside transporters, hENT1–2), and SLC22 (organic cation transporters, hOCT1–3 and hOCTN1–2). Both hENTs and hCNT2 were abundant in primary lymphocytes, with a preferential activity of hENT1. A significant up-regulation in hENTs expression (100-fold) and activity (30-fold) was seen under stimulation of primary T lymphocytes. In contrast, monocytes, monocyte-derived macrophages (MDMs), and immature monocyte-derived dendritic cells predominantly expressed hCNT3, a functional transporter in MDMs. Finally, in immune cells, hOCTs showed a more heterogeneous expression profile and a lower activity than human nucleoside transporters (hNTs), although up-regulation of hOCTs also occurred upon lymphocyte activation. Overall, the expression and activity of most of the studied transporters emphasize their relevance in relation to anti-HIV and anticancer therapies. The identification of the transporter involved in each specific drug uptake in immune cells could help to optimize pharmacological therapeutic responses.

Most of the drugs currently used in the treatment of the human immunodeficiency virus type-1 (HIV-1) infection and in anticancer therapies need to cross cell membranes to develop their pharmacological activity. Thus, host factors, including drug transporters, drug-metabolizing, and detoxifying enzymes, might affect intracellular drug concentrations, limiting the ability of drug regimens to inhibit HIV-1 replication or proliferation of cancerous cells (Evans and Relling, 1999; Fridland et al., 2000). However, little is known regarding drug uptake transporters expression and activity in immune cells.

During the past few years, many membrane proteins have been identified as putative transport systems that regulate cell entry of many antiviral and anticancer drugs. The best known uptake transporters belong to the SLC29 and SLC28 gene families, which correspond to human equilibrative nucleoside transporters (hENTs) and human concentrative nucleoside transporters (hCNTs), respectively. The best characterized isoforms of hENTs, hENT1 and hENT2, are low-affinity (Km 50–700 μM), bidirectional, and equilibrative nucleoside transporters that are widely distributed in human tissues (Baldwin et al., 2004). hENT1 has been described as transporter of the anti-HIV nucleoside analogs zalcitabine (ddC) and didanosine (ddI), whereas hENT2 is considered a low-affinity azidothymidine (AZT) transporter (Baldwin et al., 2004). Both hENTs mediate the entry of most of the drugs used in anticancer treatment: fludarabine, cladribine, cytarabine, and gemcitabine (Zhang et al., 2007). In relation to hCNTs, the sodium-dependent nucleoside family with a high substrate affinity (Km 1–50 μM), three isoforms have been described with differential substrate selectivity; whereas hCNT1 can transport pyrimidines, hCNT2 is a purine-preferring transporter that can also transport uridine and hCNT3 is a broad selective nucleoside transporter (Gray et al., 2004). Although hCNT1 has been described as the uptake transporter for ddI and stavudine (d4T), we and others (Cano-Soldado et al., 2004; Chang et al., 2004) have demonstrated that the lack of the 3′-OH group in the ribose ring of the nucleoside analogs structure causes a substantial reduction in hCNT1/2 and hENT1 affinity for these drugs. Nevertheless, hCNT3 seems to be a good transporter for AZT, ddC, ddI, and some important anticancer drugs, such as fludarabine, gemcitabine, and 5′-DFUR (Pastor-Anglada et al., 2005; Hu et al., 2006; Errasti-Murugarren et al., 2007).

On the other hand, some members of SLC22 gene family that comprises human organic anion transporters (hOATs) and organic cation/zwitterion transporters (hOCTs and hOCTNs, respectively) have also been studied as uptake transporters of different antiretroviral and anticancer drug families in secretion and reabsorption tissues (Aiba et al., 1995; Zhang et al., 2000; Lee and Kim, 2004). Four hOATs members (hOAT1–4) have been cloned and identified so far, and their substrates include cyclic nucleotides, prostaglandins, uric acid, and different types of drugs (including antiviral and anticancer nucleosides) (Koepsell and Endou, 2004). Nevertheless, we have recently demonstrated that these transporters are not expressed in T-lymphocytes or macrophages (Purcet et al., 2006). With regard to hOCTs, they mediate the first step in hepatic or renal excretion of many cationic and amphiphilic drugs and affect interstitial concentrations of endogenous compounds, drugs, and xenobiotics in a variety of tissues (Koepsell et al., 2007). hOCT1 and hOCT2 mediate the uptake of oxaliplatin, a platinum-derived drug used in cancer (Zhang et al., 2006). hOCT1 also transports imatinib, and its expression is important in the clinical response of this drug broadly used in chronic myeloid leukemia (White et al., 2006; Wang et al., 2007). Moreover, some hOCTs could be involved in the uptake of anti-HIV drugs as it has been described for AZT and lamivudine (3TC) uptake by rat OCTs and a nondetermined type of organic cation-proton exchanger (Aiba et al., 1995; Hong et al., 2001; Leung and Bendayan, 2001). Finally, hOCTN1 and hOCTN2, belonging to organic cation/zwitterion transporters, are linked to H+ and Na+ cotransport, respectively. hOCTN1 mainly transports cationic molecules, whereas hOCTN2 is a high-affinity transporter for l-carnitine (Koepsell and Endou, 2004).

It is interesting to note that, whereas in secretory tissues only carrier-dependent mechanisms are described in the uptake of anti-HIV drugs, the few studies published in leukocytes suggest nonfacilitated diffusion mechanisms (Mahony et al., 2004). Nevertheless, we have recently shown that approximately 60% AZT entry in the T-cell line Molt-4 involves a facilitated mechanism (Purcet et al., 2006).

In the present study, we have assessed the mRNA levels, protein expression profile, and physiological activity of membrane transporters codified by SLC28, SLC29, and SLC22 genes in target cells for HIV-1 infection and some leukemias, including peripheral blood mononuclear cells (PBMCs), T-lymphocytes (lymphoblastoid T-cells and purified CD4+ T-cells), monocytes, macrophages, and dendritic cells.

Materials and Methods

Tissue Specimens from Human Donors. PBMC were obtained from healthy human volunteers, and a lysate of normal kidney tissue was obtained from renal biopsy. The institutional ethics review board approved the study in adherence to the Helsinki Declaration.

Reagents. Uridine ([5,6-3H], 37 Ci/mmol), l-carnitine hydrochloride ([H3C-3 H], 83 Ci/mmol), and mannitol (d-[1-14C], 59 mCi/mmol) were purchased from Amersham Biosciences (Buckinghamshire, UK). Cytidine ([5-3H(N)], 22 Ci/mmol) was purchased from Moravek Biochemicals (Brea, CA), and N-methyl-4-phenylpyridinium (MPP+) ([H3C-3H], 85 Ci/mmol) was purchased from American Radiolabeled Chemicals (St. Louis, MO).

The inhibitors of nucleoside transporters nitrobenzylthioinosine (NBTI), dipyridamole (DIP), and substrates and inhibitors of organic cation transport ranitidine (Rani), ergothioneine (ET), and D-22 were obtained from Sigma-Aldrich (St. Louis, MO).

Cell Culture. Six different lymphoblastoid cell lines (MT2, MT4, Molt-4/CCR5, Hut78, A3F7, and CEM13) were used to quantify nucleoside and organic cation transporter expression by real-time PCR. The T-cell leukemia cell lines MT2 and MT4 were obtained from Dr. D. Richman; T4-lymphoblastoid human cell line Molt-4/CCR5 was from Dr. M. Baba; and Hut78 human cutaneous T-cell lymphoma cell line was from Dr. A. F. Gazdar, all of these through the National Institutes of Health AIDS Research and Reference Reagent Program. The T4-cell line A3.01/CCR5 F7 (abbreviated here as A3F7) was obtained from Dr. Q. Sattentau through the Centre for AIDS Reagents. Clone 13 (CEM13), which was derived from the T4-cell line CEM, was kindly donated by Dr. L. Montagnier from Institute Pasteur (Paris, France). Cells were routinely cultured in RPMI 1640 culture medium supplemented with 10% heat-inactivated fetal bovine serum, 2 mM l-glutamine, and 100 U/ml penicillin and 100 mg/ml streptomycin (Invitrogen, Paisley, UK), with the exception of the A3F7 cells, which were also supplemented with 1 mg/ml G-418 (Invitrogen) and maintained at 37°C in a humidified atmosphere containing 5% CO2.

PBMCs were isolated from HIV-1-seronegative donors by Ficoll-Hypaque density gradient centrifugation of heparin-treated venous blood. CD4+ T-cells were isolated from PBMC by negative selection with the CD4+ Isolation Kit II (Miltenyi Biotec, Bergisch Gladbach, Germany), and all of the isolated populations had a high purity (>95%). Cells were stimulated for 3 days by adding 3 μg/ml phytohemagglutinin (PHA; Invitrogen) and 10 IU/ml interleukin-2 (IL-2; Roche, Basel, Switzerland). Monocytes were isolated with a high purity (>97%) from PBMC with CD14+-positive selection magnetic beads (Miltenyi Biotec). We differentiated macrophages by culturing monocyte populations (0.8 × 106 cells/ml) with 100 ng/ml macrophage-colony-stimulating factor (Peprotech, London, UK). We maintained them in culture for 5 to 6 days replacing medium at day 3.

To obtain immature dendritic cells populations, we cultured monocytes (0.8 × 106 cells/ml) during 7 days with 1000 IU/ml granulocyte/macrophage colony-stimulating factor (GM-CSF) and 1000 IU/ml IL-4 (R&D Systems, Minneapolis, MN). Medium was replaced every 2 to 3 days adding fresh GM-CSF and IL-4. To obtain mature dendritic cells population, 100 ng/ml lipopolysaccharide (LPS; Sigma-Aldrich) was added at day 5, and the culture was maintained until day 7.

All primary cells were maintained in RPMI 1640 medium supplemented with 2 mM l-glutamine, 20% fetal bovine serum (10% for macrophages), and 100 U/ml penicillin and 100 mg/ml streptomycin (Invitrogen).

Quantitative Real-Time Reverse Transcriptase-PCR Analysis. Total RNA was isolated from different lymphoblastoid cell lines, CD4+ lymphocytes, PBMCs, macrophages, and kidney lysates using RNeasy Mini Kit or RNeasy Micro Kit (QIAGEN, Barcelona, Spain). RNA was treated with DNase I from RNase-free DNase set (QIAGEN) to eliminate contamination of DNA. In total, 1 μg of RNA was retrotranscribed to cDNA using the TaqMan reverse transcription reagents (including Multiscribe reverse transcriptase and random hexamers) as described by the manufacturer (Applied Biosystems, Foster City, CA). Real-time quantitative reverse transcriptase-PCR analysis of hENT1, hENT2, hCNT1, hCNT2, hCNT3, and human β-glucuronidase mRNA was performed as described previously (Molina-Arcas et al., 2003). The mRNA expression of hOCT1, hOCT2, hOCT3, hOCTN1, and hOCTN2 was assessed using the commercial Gene Expression Assays (Applied Biosystems). Absolute quantification of gene expression was performed by using DNA plasmids containing each of the analyzed transporters to construct standard curves based on serial dilutions of the plasmids. Standard curves were optimized in terms of correlation, slope, and efficiency and were run in duplicate simultaneously with the samples in the ABI Prism 7000 (Applied Biosystems). The threshold cycle (CT) is defined as the cycle number at which the fluorescence corresponding to the amplified PCR product is detected. The standard curves allowed us to correlate CT values of the samples with the mRNA copy number of each gene per microgram of total RNA.

Immunocytochemistry. Cytocentrifuge preparations (cytospins) of PBMCs or monocyte-derived macrophages (MDMs) cultured on poly-l-lysine-treated coverslips (Nunc, Roskilde, Denmark) were air-dried overnight and fixed in 100% acetone for 10 min. Slides were incubated with polyclonal antibodies diluted in Tris-buffered saline (TBS) containing 2% normal human serum or fetal bovine serum to block unspecific binding. Primary rabbit polyclonal antibodies anti-hCNT2, -hCNT3, -hENT1, and -hENT2 were produced in our laboratories (Mata et al., 2001; Farre et al., 2004). The newly generated anti-hCNT2 and anti-hCNT3 antibodies were characterized by competing them with an excess of the purified peptide used to immunize the rabbit, as well as by analyzing hCNT2 and hCNT3 expression in transfected cells expressing these particular transporter proteins (data not shown). For cytospins of PBMCs or MDMs cultured on coverslips, all primary antibodies were detected using a polyclonal goat anti-rabbit-conjugated antibody (Cultek, Barcelona, Spain) followed by development with liquid DAB+ substrate (DakoCytomation, Madrid, Spain) as described by the manufacturer. After washing with TBS and fixing with 4% paraformaldehyde, samples were incubated in Nuclear Fast Red (DakoCytomation) for PBMCs or hematoxylin (DakoCytomation) for MDMs for 5 min to counterstain nuclei, washed in TBS, dehydrated with ethanol and xylene, and mounted with Distyrene-Plasticizer-Xylene medium (Sigma-Aldrich). For negative controls, slides were stained with isotype-matched human or rat antibodies. Images were collected at 400× using an Axioskop Microscope (Carl Zeiss, Barcelona, Spain) and a Leica 330 DFX camera and the software Leica Cam (Leica, Barcelona, Spain).

Western Blot Analysis. HEK293T, a human embryonic kidney cell line (15 × 106), and primary cultures of PBMCs, PHA-stimulated PBMCs, CD4+ T-cells, PHA-stimulated CD4+ T-cells, monocytes, MDMs, immature monocyte-derived dendritic cells (iMDDCs), and mature MDDCs (mMDDCs) (20 × 106 in all cases) were harvested, washed with phosphate-buffered saline, and lysed in chilled hypotonic buffer (20 mM Tris-HCl, 140 mM NaCl, 10% glycerol, 1% Nonidet P-40, and 2 mM EDTA, pH 8.0, freshly supplemented with a cocktail of protease inhibitors and 100 mM phenylmethylsulfonyl fluoride (Sigma-Aldrich). Cell debris was removed by centrifugation, and quantification of protein concentration was performed by bicinchoninic acid (Pierce, Madrid, Spain). Protein samples were run using 4 to 12% ν-polyacrylamide gel electrophoresis gels (Invitrogen) and blotted onto nitrocellulose membranes by using iBlot Dry Blotting System as described by the manufacturer (Invitrogen). Membranes were blocked with 0.5% phosphate-buffered saline-Tween 20, 5% nonfat milk (blocking solution) for 4 h at room temperature and incubated overnight with rabbit polyclonal antibodies against rat OCT1 and OCT3 or against mouse OCTN1 (diluted 1/100 in blocking solution; Alpha Diagnostic, San Antonio, TX), all cross-reacting with human orthologs or mouse monoclonal antibody against actin (1/3000 in blocking solution; Chemicon, Barcelona, Spain) at 4°C. After washing, membranes were incubated with goat anti-rabbit horseradish peroxidase (1/2000 in blocking solution; Cultek) or rabbit anti-mouse (Invitrogen) for 45 min at room temperature and then revealed with ECL-Plus solution (Amersham Biosciences). Images were collected using Kodak Gel Logic 440 Imaging System and Kodak Molecular Imaging Software (Kodak, Barcelona, Spain).

Uptake Measurements. The uptake of uridine, the organic cation model (MPP+), and l-carnitine into PBMCs and PHA-stimulated PBMCs was measured using a rapid filtration method as described previously (Molina-Arcas et al., 2003). Having previously determined in time-course experiments with radiolabeled substrates that the uptake linearity of the substrates was maintained at least until 5 min for uridine and MPP+ and until 10 min for l-carnitine in those cells, PBMCs were incubated for 2 (uridine and MPP+) or 5 min (l-carnitine) in an uptake buffer containing 137 mM NaCl or 137 mM choline chloride plus 5.4 mM KCl, 1.8 mM CaCl2, 1.2 mM MgSO4, and 10 mM HEPES, pH 7.4, supplemented with radiolabeled uridine or MPP+, or l-carnitine and d-[1-14C]mannitol as an extracellular marker and inhibitor of both nucleoside or organic cation transport when necessary. Substrates were routinely used at a concentration of 1 μM as done previously in the case of uridine (Purcet et al., 2006) and based on kinetic parameters (Km and Vmax) for MPP+ and l-carnitine as reported previously (Koepsell and Endou, 2004; Koepsell et al., 2007).

Transport studies in MDMs were carried out on 24-well plates as described previously (Soler et al., 2003) by incubating semiconfluent cell monolayers for 10 min in an uptake buffer of choline chloride or sodium chloride supplemented with radiolabeled uridine or cytidine or the organic cation model (MPP+) at a final concentration of 1 μM in all cases. The uptake was measured either in the presence or absence of the equilibrative nucleoside inhibitors NBTI (1 μM) or dipyridamole (10 μM) in nucleoside uptake experiments and the organic cation inhibitor D-22 (20 μM) or the high-affinity competitive substrates Rani and ET at 1 mM in organic cation uptake measurements. Incubations were stopped by rapid aspiration of the uptake buffer, followed by immediate washing in ice-cold stop solution (137 mM NaCl, 10 mM HEPES, pH 7.4).

Statistical Analysis. The unpaired and paired Student's t tests were used for the statistical comparison of experimental data. These analyses were carried out using GraphPad Prism version 4.0 software (GraphPad Software Inc., San Diego, CA).

Results

mRNA Expression of Nucleoside and Organic Cation Transporters. Because of the importance of the SLC28 and SLC29 families in nucleoside-derived drug transport, especially those used in cancer and HIV treatment, we first focused our attention on the expression of hNTs in lymphoblastoid T-cell lines by examining the mRNA levels of both concentrative (hCNT1–3) and equilibrative nucleoside transporters (hENT1–2) (Fig. 1A). hCNT2 was the most expressed concentrative isoform in all of the cell lines tested, being more expressed in MT4, whereas hCNT1 and hCNT3 showed either low or negligible expression. Both hENTs showed a very high expression (between 105 and 107 mRNA copy number/microgram of RNA).

  Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

hCNT and hENT mRNA expression in T-cell lines, primary lymphocytes, monocytes, MDMs, and MDDCs cultures. A, mRNA expression of hNTs in six different T-lymphoblastoid cell lines (MT2, MT4, Molt-4, CEM13, A3F7, and Hut78). B, mRNA expression of hNTs in nonstimulated PBMCs (PBMCs) or purified CD4+ T-lymphocytes (CD4+) and 3 days of PHA-stimulated PBMCs (Stim PBMCs) or CD4+ T-lymphocytes (Stim CD4+). Statistical significance between PBMCs/CD4+ and Stim PBMCs/CD4+ (*, p < 0.05; **, p < 0.01; ***, p < 0.001) and between PBMCs and CD4+ (#, p < 0.05 and ##, p < 0.01) was assessed by paired Student's t test. C, mRNA expression of monocytes, MDM, and monocyte-derived dendritic cells, both iMDDC and mMDDC. Statistical significance between monocytes and MDMs/iMDDCs (*, p < 0.05; **, p < 0.01) and between iMDDCs and mMDDCs (##, p < 0.01) was assessed by paired Student's t test. The data represent the mean ± S.E.M. of at least four independent experiments performed in duplicate. DL represents the theoretical detection limit of the technique.

We then studied the mRNA expression of hNTs in PBMCs and in purified CD4+ T-cells either in basal conditions or after PHA stimulation of these cells. As shown in Fig. 1B, similar to T-lymphoblastoid cells, hCNT2 was the only concentrative nucleoside transporter substantially expressed in PBMCs and CD4+ T-cells. Under PHA stimulation, all hCNTs appeared to up-regulate their expression, although only the increase in hCNT2 transcripts in PBMCs was statistically significant (p = 0.005). hENT-related mRNA levels were similar to those of hCNT2 in nonstimulated PBMCs (approximately 103 copies/μg of RNA). In that case, the PHA stimulation caused a statistically significant up-regulation in both PBMCs and CD4+ T-cells, with hENT1 being the most up-regulated transporter (p < 0.01 and 0.005, respectively). Moreover, the greatest difference between PBMCs and CD4+ T-cells, when looking at basal transporter expression, was associated with hENTs, both showing higher mRNA-related levels in PBMCs (p = 0.005 for hENT1 and p = 0.035 for hENT2) than in CD4+ T-cells.

To further study hNT expression patterns in immune cells, we determined the mRNA levels for SLC28 and SLC29 gene transporter families in monocytes, MDMs and MDDCs. In contrast to PBMCs and CD4+ T-cells, monocytes showed a remarkably higher expression of hCNT3. Moreover, monocyte differentiation to MDMs and iMDDCs produced a significant increase of hCNT3 expression (p = 0.03 and 0.005, respectively). As for T-cell lines and primary lymphocytes, hCNT2 and both hENTs isoforms showed a notable expression, with hENT1 being the most expressed transporter. The high expression of hENT1 and hENT2 in monocytes (approximately 105 and 104 copies mRNA/μg RNA, respectively) could explain the higher mRNA amounts of those transporters in PBMCs compared to CD4+ T lymphocytes.

Focusing on SLC22 gene family, hOATs were ruled out from this study because they are were expressed in Molt-4, but not in primary T-cells or in macrophages (Purcet et al., 2006). Even though we aimed at studying the expression of hOCTs and hOCTNs, which also belong to the SLC22 gene family and are known to be involved in antiviral and anticancer drug uptake (Koepsell et al., 2007). Organic cation transporters showed lower expression and a more heterogeneous expression profile than nucleoside transporters in T-cell lines (Fig. 2A). It is noteworthy that none of the cell lines tested had detectable hOCT2 mRNA levels.

In nonstimulated primary PBMCs and CD4+ T-cell cultures (Fig. 2B), hOCT1 and hOCTN2 had nearly the same mRNA levels in both populations, whereas CD4+ T-cells showed a significantly lower hOCT3 and hOCTN1 mRNA expression than PBMCs (p = 0.002 for both transporters). As seen in T-cell lines, hOCT2 mRNA was not found in any of primary lymphocytes tested. Under PHA stimulation, all hOCTs and hOCTNs previously expressed underwent a significant up-regulation. The greatest up-regulation in PBMCs and CD4+ T-cells was seen for hOCT3 and hOCTN2 (p < 0.01 for hOCT3 and p < 0.001 for hOCTN1 in both cell types).

Regarding monocytes/macrophage lineage primary cultures (Fig. 2C), hOCTs and hOCTNs showed the most heterogeneous and cell type depending on mRNA expression pattern. In regard to monocytes, they showed the highest mRNA expression of hOCT3 and hOCTN1. The differentiation of monocytes to macrophages with macrophage-colony-stimulating factor or to dendritic cells with GM-CSF/IL-4 seems to have an antagonistic effect on hOCT and hOCTN expression. Whereas for hOCT1 and hOCTN2 transcripts, there is a statistically significant up-regulation (p < 0.05 and 0.001, respectively), hOCT3 and hOCTN1 mRNA copy numbers decrease in a significant manner. When iMDDCs were matured with LPS, the expression of hOCTN1 increased respect to MDMs.

  Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

hOCT and hOCTN mRNA expression in T-cell lines, primarily lymphocytes, monocytes, MDMs, and MDDCs cultures. A, mRNA expression of hOCTs and hOCTNs in six different T-lymphoblastoid cell lines (MT2, MT4, Molt-4, CEM13, A3F7, and Hut78). B, mRNA expression of hOCTs and hOCTNs in nonstimulated PBMCs (PBMCs) or purified CD4+ T-lymphocytes (CD4+) and 3 days of PHA-stimulated PBMCs (Stim PBMCs) or CD4+ T-lymphocytes (Stim CD4+). Statistical significance between PBMCs/CD4+ and Stim PBMCs/CD4+ (*, p < 0.05; **, p < 0.01; ***, p < 0.001) and between PBMCs and CD4+ (##, p < 0.01) was assessed by paired Student's t test. C, mRNA expression of monocytes, MDM, and iMDDC and mMDDC. Statistical significance between monocytes and MDMs/iMDDCs/mMDDCs (*, p < 0.05; **, p < 0.01; ***, p < 0.001) was assessed by paired Student's t test. The data represent the mean ± S.E.M. of at least six independent experiments performed in duplicate. DL represents the theoretical detection limit of the technique.

Protein Expression of hNTs and hOCT/Ns. Having studied mRNA expression of hNTs and hOCT/Ns and knowing that there might not be systematic correlation between mRNA and protein expression (Molina-Arcas et al., 2005), we wanted to assess the protein expression profile in PBMCs and MDMs and the effect that PHA would cause in the transporter expression in PBMCs. As shown in Fig. 3, we analyzed both hCNTs and hENTs expression in cytospined PBMCs by immunocytochemistry developed by horseradish peroxidase (HRP). In those cells, we found that mRNA and protein had the same expression profile, nonstimulated PBMCs expressing hCNT2 and hENT1 and 2 proteins, whereas hCNT1 and hCNT3 were not detected (data not shown). Moreover, the expression of hCNT2 and both hENTs in PHA-stimulated PBMCs was increased, with hENT2 up-regulation being the highest. Immunocytochemistry performed in MDMs cultured on coverslips showed a high expression of hCNT2, hCNT3, and both hENT isoforms (Fig. 4) and a negligible expression of hCNT1 (data not shown).

  Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Immunocytochemistry of hCNT2 and hENTs in nonstimulated and PHA-stimulated PBMC cytospins. Cytocentrifuge preparations of PBMCs (nonstim PBMCs) or PHA-stimulated PBMCs (PHA-stim PBMCs) were fixed, and primary rabbit-specific polyclonal anti-hCNT2, -hENT1, and -hENT2 antibodies were used to detect protein expression. A secondary conjugate and the development of activity with DAB+ substrate (brown) were used for the detection of primary antibody binding. Nuclei were counterstained with Nuclear Fast Red (light pink). For negative controls, slides were stained with isotype-matched (IgG) human antibodies. Intense brown circular stained cells are residual erythrocytes (with a high peroxidase content in their membranes) present in the cytospin preparations.

We analyzed the protein expression of hOCT1, hOCT2, hOCT3, and hOCTN1 in nonstimulated and PHA-stimulated PBMCs, purified CD4+ T-cells, and monocytes MDMs and MDDCs by Western blot (Fig. 5). In agreement with mRNA expression levels, we observed an increase in hOCT1, hOCT3, and hOCTN1 protein amounts following PHA stimulation of PBMC and CD4+ T-cells. hOCT2 protein was not detected in any of the blood cells studied (data not shown). hOCT1 protein expression could not be detected in monocytes, macrophages, and MDDCs, even though mRNA levels were high. In relation to hOCT3, showing less associated mRNA levels in MDMs and MDDCs, a higher protein expression in monocytes and macrophages than in MDDCs was observed. hOCTN1 showed a notable expression in MDMs.

  Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Immunocytochemistry of hCNT3 and hENTs in MDMs cultured on coverslips. Monocyte-derived macrophages were cultured in poly-l-lysine-treated coverslips and fixed. Primary rabbit specific polyclonal anti-hCNT3, -hENT1, and -hENT2 antibodies were used to detect protein expression. A secondary conjugate and the development of activity with DAB+ substrate (brown) were used for the detection of primary antibody binding. Nuclei were counterstained with hematoxylin (blue-violet). For negative controls, slides were stained with isotype-matched (IgG) human antibodies.

  Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Western blot analysis of hOCT1 (A), hOCT3 (B), and hOCTN1 (C) in immune cells. Lines correspond to HEK293T (293T-Ctrl-), PBMCs, PHA-stimulated PBMCs (PBMCs stim), CD4+ T-cells, PHA-stimulated CD4+ T-cells (CD4+ stim), and monocytes MDMs, iMDDCs, and mMDDCs. Actin was used as a control for protein loading.

Functional Studies of Nucleoside and Organic Cation Transport in PBMCs. We aimed at determining the physiological function of both nucleoside transporters and organic cation transporters in primary lymphocytes (in both nonstimulated and PHA-stimulated PBMCs). To accomplish this objective, we used uridine and MPP+, model substrates of hNTs and hOCTs, respectively, and we carried out cis-inhibition experiments. As shown in Fig. 6A, equilibrative uridine transport in nonstimulated PBMCs was very low (0.3–1.0 pmol/mg protein) and could be completely inhibited (p < 0.05) both by DIP and NBTI, thus confirming that all equilibrative nucleoside transport was mainly due to hENT1 activity. Under PHA stimulation, a significant up-regulation of uridine transport was seen (p = 0.004; 5.5–9.0 pmol/mg protein), and this could also be almost completely inhibited by NBTI (p = 0.001). This reflects the minimal contribution of hENT2 activity to nucleoside transport in primary lymphocytes. In addition, the Na+ component of nucleoside transport (that would be attributed to hCNT2) seemed to be residual, nearly negligible. However, it is important to remark that the PBMCs of some of the donors showed partial Na+ dependence of uridine uptake, suggesting detectable hCNT2 function.

When addressing OCT-related functional activity, we observed a relatively high percentage of transport in nonstimulated PBMCs (0.5–1.3 pmol/mg protein) (Fig. 6B) that could be related to the high lipophilicity of MPP+. Actually, MPP+ transport could not be inhibited by either Rani, D-22, or ET. When PBMCs were stimulated with PHA, MPP+ transport was increased 3-fold above basal (nontreated) cells (2.3–3.0 pmol/mg protein). A notable inhibition of MPP+ uptake could be observed with the addition of ranitidine (32.7% inhibition; p < 0.001) and D-22 (34.4%; p = 0.001). Moreover, the high-affinity hOCNT1 substrate ET could inhibit MPP+ transport significantly (24.2%, p = 0.018) but to a lesser extent than that exerted by hOCT substrates or inhibitors.

Functional Studies of l-Carnitine Transport in PBMCs. To assess the functionality of hOCTN2, a Na+-dependent high-affinity transporter for l-carnitine (Tamai et al., 2000; Duran et al., 2005), we performed uptake of l-[3H]carnitine in nonstimulated and PHA-stimulated PBMCs in the absence (choline medium) and presence of Na+ (NaCl). In the presence of Na+, we found a notable l-carnitine transport in both nonstimulated and PHA-stimulated PBMCs (Fig. 7), with the increase of transport in stimulated cells (447 ± 25 versus 765 ± 66 fmol/106 cells; p = 0.01) being statistically significant. It is surprising that we also found a high transport rate of l-carnitine in the absence of Na+ both in nonstimulated PBMCs (365 ± 6 fmol/106 cells; 82% transport in NaCl) and in PHA-stimulated PBMCs (516 ± 87 fmol/106 cells; 67% transport in NaCl). Finally, the difference between the transport in the absence and presence of Na+ in nonstimulated PBMCs was statistically significant (365 ± 6 versus 447 ± 25 fmols/106 cells; p = 0.03).

Functional Studies of Nucleoside and Organic Cation Transport in MDMs. To further study the function of drug uptake transporters in immune cells, we investigated the importance of hNTs and hOCTs in MDMs. This was done in macrophages due to their importance in inflammatory processes during cancer and viral infections and their role as important latent reservoirs for HIV-1 (Aquaro et al., 2002). As we can see in Fig. 8A, normalizing data to 100% transport with uridine transport in Na+ medium (with both hCNTs and hENTs being operative), the hCNT-associated transport of uridine (57.5%) is much higher than the one found in PBMCs (3.4%; nearly negligible). Uridine transport associated with hENT-type activities could be significantly inhibited by both the specific hENT1-inhibitor NBTI (p < 0.05) and the generic hENT inhibitor DIP (p < 0.05). This agrees with data obtained in PBMCs, where hENT2 function was absent in spite of the high mRNA levels. Moreover, the difference between uridine transport rates measured in a choline chloride (when only hENTs are functional) and a sodium chloride medium (with hCNTs and hENTs actively transporting) was statistically significant (9.6 ± 1.6 versus 27.6 ± 10.4 pmol/mg protein; p = 0.003), a feature that highlights the relevance of hCNTs in macrophages.

  Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

Uridine and MPP+ transport in nonstimulated and PHA-stimulated PBMCs. A, the equilibrative (CHO; white bars), NBTI-inhibited (light gray bars), and DIP-inhibited uptake (dark gray bars); the amount of concentrative and equilibrative (Na+; black bars); and the amount of concentrative and equilibrative DIP-inhibited uptake (white-grated bars) of 1 μM[3H]uridine were measured for 2 min either in a choline chloride or NaCl medium in PBMC cells from at least four independent healthy donors (performed in triplicate). B, the Na+-independent uptake of 1 μM [3H]MPP+ was measured for 2 min in a choline chloride medium in absence (Ctrl; white bars) or in the presence of the hOCT/N substrates Rani (1 mM; light gray bars) and ET (1 mM; dark gray bars), and hOCT inhibitor D-22 (20 μM; black bars) in PBMCs from at least four independent healthy donors (performed in triplicate). The experiments were carried out in both nonstimulated (Nonstim) and 3 days of PHA-stimulated PBMCs (PHA-stim). Results are expressed as the mean ± S.E.M. Statistical significance between CHO/control versus different conditions of transport and between nonstim CHO/control versus stimulated CHO/control was assessed by paired Student's t test (*, p < 0.05; **, p < 0.01; ***, p < 0.001).

To elucidate whether this Na+-coupled transport activity was related to hCNT3 expression in MDMs, guanosine inhibition of cytidine uptake was monitored, because hCNT3 is known to transport both purines and pyrimidines. Figure 7B shows a representative cytidine transport experiment in MDMs for one blood donor. Actually, high variability of cytidine transport rates among donors was observed (1.7–7.0 pmol/mg protein in choline chloride and 5.7–14.0 pmol/mg protein in NaCl medium). Results showed that concentrative cytidine uptake could be inhibited by 1 mM guanosine, thus revealing hCNT3-related activity. Cross-inhibition of pyrimidine nucleoside transport by purines was also highly variable in donors (from 40 to 95%).

Finally, we studied MPP+ transport processes in MDMs (Fig. 8C). As for PHA-stimulated PBMCs, MPP+ basal transport (12.7 ± 4.2 pmol/mg protein) could be inhibited by the hOCT substrate ranitidine (34.4% inhibition; p < 0.01), the hOCTN1 substrate ergothioneine (27.9%; p < 0.01), and the hOCT inhibitor D-22 (57.3%; p < 0.05). This reflects the importance of hOCT and hOCTN function in macrophages and corroborates our results of mRNA and protein expression.

Discussion

The expression of drug transporters is relatively well studied in secretory and reabsorptive tissues in relation to toxicity, drug-drug interactions, and drug resistance in antineoplasic and antiviral therapies (Lee and Kim, 2004; McRae et al., 2006; Koepsell et al., 2007). Moreover, in relation to peripheral blood cells exists a broad knowledge about the expression and activity of efflux transporters (Kock et al., 2007). However, little is known regarding the expression profile and activity of uptake transporters in human leukocytes within the context of anticancer and antiretroviral therapies. In the present study, we have assessed the mRNA levels, protein expression profile, and activity of drug uptake transporters involved in anti-HIV and antineoplasic drug uptake in cells of the immune system. Taking into account that HIV retrotranscriptase inhibitors and anticancer drugs are mostly nucleoside derivatives, our first focus was on the nucleoside transporters from the SLC29 and SLC28 families. Likewise, members of the SLC22 family (OATs and OCTs) have also been described as drug transporters in other tissues. We had demonstrated that OATs were not expressed in immune system cells (Purcet et al., 2006); thus, we have only analyzed OCT transporters, which have not been previously characterized in immune system cells.

First, we analyzed the mRNA and protein expression of hCNTs and hENTs. In both T-cell lines and primary lymphocytes (PBMCs and CD4+ T-cells), hENTs were the most expressed transporters, whereas hCNT2 was the only sodium-dependent nucleoside transporter remarkably expressed. These results are in agreement with the previously described expression profile of nucleoside transporters in primary lymphocytes (Smith et al., 1989; Wiley et al., 1989; Molina-Arcas et al., 2003; Molina-Arcas et al., 2005). Second, PHA stimulation significantly up-regulated hENTs and hCNT2 in PBMCs and CD4+ T-cells. A similar up-regulation had been described for CNT1 and CNT2 upon stimulation of murine bone marrow macrophages with interferon-γ-α/β/γ, tumor necrosis factor-α, or LPS (Soler et al., 2001, 2003). In contrast, all of these stimuli slightly down-regulated the expression of ENT1 in murine macrophages (Soler et al., 2001), suggesting that the up-regulatory effect of PHA on hENTs might act through a different pathway. Moreover, the PHA-induced up-regulation of hENT1 is in agreement with the 30-fold increase in the number of hENT1 molecules previously found in peripheral blood lymphocytes cultured within 48 h with the T-cell mitogen (Smith et al., 1989). It is also important to remark that PHA stimulation of PBMCs and CD4+ T-cells enhances hENT expression to a level similar to that reported in T-acute lymphoblastic leukemia. In fact, an increased proliferative rate of acute myeloid leukemia or lymphoma is associated with higher numbers of nucleoside transporters in the cell membrane (Wiley et al., 1989). As in the case of T-lymphocytes and T-cell lines, significant mRNA levels were also seen for hCNT2, hENT1, and hENT2 in monocyte/macrophage lineage cells. However, monocyte/macrophage lineage cells expressed important quantities of hCNT3, whereas T-lymphocytes did not. Thus, iMDDC and MDM showed the highest number of hCNT3 mRNA copies even though monocytes and mMDDCs also showed greater expression than PBMCs or CD4+ T-cells. The abundance of hCNT3 protein in macrophages was also confirmed by immunocytochemistry. Of note, the differential expression profile of this transporter in monocyte/macrophage lineage cells is a novel observation in this field.

  Fig. 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 7.

l-Carnitine transport in nonstimulated and PHA-stimulated PBMCs in the presence and absence of sodium. The Na+-independent (CHO; white bars) and the Na+-dependent uptake (Na+; black bars) of 1 μM l-[3H]carnitine was measured for 5 min in nonstimulated (Nonstim) and PHA-stimulated PBMCs (PHA-stim) from three independent healthy donors (performed in triplicate). Results are expressed as the mean ± S.E.M. Statistical significance between CHO versus Na+ in nonstimulated PBMCs and PHA-stimulated PBMCs was assessed by paired Student's t test (*, p < 0.05; **, p < 0.01).

  Fig. 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 8.

Uridine, cytidine, and MPP+ transport in MDMs. A, the equilibrative (CHO; white bars), NBTI-inhibited (light gray bars), and DIP-inhibited uptake (dark gray bars) and the amount of concentrative and equilibrative uptake (Na+, black bars) of 1 μM[3H]uridine were measured during 2 min either in a choline chloride or NaCl medium in MDMs from four independent healthy donors (performed in triplicate). B, the uptake of 1 μM [3H]cytidine during 2 min in choline chloride medium in absence (ENTs; white bar) or presence of 1 mM guanosine (ENTs + Guo 1 mM; light gray bar) and the Na+-dependent cytidine transport in absence (CNTs; dark gray bar) or presence of 1 mM guanosine (CNTs + Guo 1 mM; black bar) is shown. A representative experiment (performed in triplicate) from one healthy donor is shown. C, the Na+-independent uptake of 1 μM[3H]MPP+ was measured for 10 min in a choline chloride medium absence (Ctrl; white bar) and the presence of hOCT/N substrates Rani (1 mM; light gray bar) and ET (1 mM; dark gray bar) and the inhibitor of hOCT D-22 (20 μM; black bar) in MDMs from four independent healthy donors (performed in triplicate). Results are expressed as the mean ± S.E.M. of the four independent donors or mean ± S.E.M. from one donor in B. In A and C, statistical significance between CHO/Ctrl versus different conditions of transport was assessed by paired Student's t test (*, p < 0.05; **, p < 0.01; ***, p < 0.001).

With regard to hNT function, uridine transport in PBMCs revealed a preferential role of hENT1. In contrast, there was a minimal contribution of sodium-dependent transporters (only hCNT2 was expressed) and a nearly negligible role of hENT2 in spite of its high and ubiquitous expression, as suggested by the complete inhibition of uridine transport in the presence of NBTI and DIP. It is important to point out that two mRNA splice variants resulting in nonfunctional transporters have been described previously (Mangravite et al., 2003). Therefore, it is possible that the protein and mRNA found for hENT2 correspond to one of these variants. Moreover, the PHA stimulation also only resulted in greater hENT1 activity accordingly with the up-regulatory effect seen on its mRNA and protein expression, whereas hENT2 did not show an increase in activity. Finally, hCNT2 activity seemed to be so low in PHA-stimulated PBMCs, although the PBMCs of some of the specific donors showed Na+ dependence of uridine uptake. This is in agreement with previous results shown by our group in B-cells from chronic lymphocytic leukemia in which, although CNT2 expression was detected in 22 patients, Na+-dependent guanosine transport activity was only detected in 12 patients (Molina-Arcas et al., 2003).

Functional analysis of hNT in MDMs mainly paralleled that in PBMCs, thus confirming that hENT2 plays a negligible role in nucleoside transport in leukocytes despite its high level of mRNA expression. The main difference found between PBMCs and MDMs was the relevant sodium component of uridine uptake in MDMs, presumably due to the presence of hCNT3. As hypothesized, the contribution of hCNT3 function was confirmed by cytidine transport inhibition by guanosine. Because cytidine is a pyrimidine nucleoside and could only be transported by hCNT1 (not expressed in MDMs) and hCNT3, the inhibition of sodium-dependent cytidine transport by guanosine demonstrates the occurrence of hCNT3 function. This finding is noteworthy by itself because it is well documented that hCNT3 is a broadly selective, potent, and high-affinity transporter for its natural substrates and, more importantly, a suitable transporter for nucleoside analogs used in anti-HIV (such as AZT, ddC, or ddI) and in anticancer therapies (such as cladribine, gemcitabine, 5-fluorouridine, and fludarabine) (Hu et al., 2006; Errasti-Murugarren et al., 2007).

With regard to hOCTs and hOCTNs, we did the same mRNA and protein expression analysis as done for hNTs. Overall, the expression profile of hOCTs and hOCTNs was more heterogeneous with a quantitative general lower expression than hNTs. PHA stimulation of PBMCs and CD4+ T-cells increased the mRNA levels and protein of hOCTs and hOCTNs, with the exception of hOCT2, which was not expressed in any of T-cell lines or primary leukocytes studied. Similar mRNA up-regulatory effects for hOCTN1 have been shown upon cell incubation with tumor necrosis factor-α (Tokuhiro et al., 2003). This expression profile of hOCTs is identical to the one that had recently been described in basophils (Schneider et al., 2005).

The physiological role that hOCTs might play in immune cells remains to be well understood, but we believe that these transporters might be important for the regulation of inflammatory processes and, as known for hOCT3, immune response related to pro-Th2 cytokines and histamine (Schneider et al., 2005). In fact, histamine for itself has a wide variety of functions in immune regulation, including vascular endothelial growth factor production via H2 receptor stimulation, mast cell chemotaxis via H4 receptor stimulation, T-cell proliferation, and dendritic cell maturation (Jutel et al., 2002). It is noteworthy that the treatment of patients with drugs with high affinity for hOCT3 might somehow impair immune response. Moreover, hOCTs might also be involved in the regulation of membrane potential, which could be important in some drug therapies using positively charged molecules, such as corticosterone, which is taken up by hOCTs (Koepsell et al., 2007).

With regard to hOCTNs, their occurrence in the immune system has only been reported so far for the hOCTN1 isoform, which is present in human cord blood cells (CD71+ cells) and leukocytes (Grundemann et al., 2005). In monocyte/macrophage lineage cells, the expression of hOCTs and hOCTNs was notably high, with monocytes being the cells with a higher expression of hOCT3 and hOCTN1. This result agrees with the maximal expression of hOCTN1 previously found in a subset of CD14+ lymphocytes (Tokuhiro et al., 2003). Moreover, monocyte differentiation to MDMs or iMDDCs caused antagonistic effects in hOCT/Ns mRNA expression; whereas hOCT1 and hOCTN2 up-regulation was observed, hOCT3 and hOCTN1 showed significant down-regulation. Furthermore, LPS maturation of iMDDCs also had an antagonistic effect on different iMDDCs hOCT/N expression. The physiological basis for this differential effect remains unexplained. Protein expression in monocyte/macrophages lineage cells could not be found for hOCT1. Nevertheless, hOCT3 showed a high protein expression in monocytes and macrophages and a lower expression in MDDCs (as seen for mRNA levels). Finally, hOCTN1 protein was highly expressed in MDMs.

The activity of hOCTs, analyzed by MPP+ transport, was lower in PBMCs with respect to the hNTs, probably because of the higher lipophilicity of organic cations and therefore higher rates of simple diffusion, even though in PHA-stimulated PBMCs, the MPP+ transport could be partially inhibited by the competing substrates ranitidine and ET, as well as by the hOCTs inhibitor D-22. In addition, because hOCTN1 is a low-affinity transporter for MPP+ (Koepsell et al., 2007) and ET is only transported by hOCTN1 with a high affinity (Grundemann et al., 2005), the inhibition seen with ET, together with the mRNA expression profile, is direct evidence that hOCTN1 is an active transporter in stimulated PBMCs. Finally, the Na+-coupled l-carnitine transport found in nonstimulated and PHA-stimulated PBMCs confirms that hOCTN2 is active in those cells. The l-carnitine uptake in the absence of Na+, which was also found, would be attributed to the recently described transporter OCTN3, which seems to be a Na+-independent l-carnitine transporter highly expressed in enterocyte basolateral membrane (Duran et al., 2005). For MDMs, we found a similar activity pattern than that for PHA-stimulated PBMCs; the substrates ranitidine and ET significantly inhibited MPP+ transport, whereas D-22 did it to a lesser extent. As far as we know, these results in PBMCs and MDMs are the first evidence of hOCT and hOCTN activity studied in primary leukocytes.

In conclusion, all of these data strengthen the importance of drug transporters associated to cell entry of hydrophilic drugs used in anticancer and antiviral therapies in immune cells. Of note, we have described the abundant presence of both hNTs and hOCTs in primary lymphocytes, monocytes, macrophages, and dendritic cells and the high activity of both transporter types in PHA-stimulated PBMC and CD4+ T-cells and MDMs. Moreover, further studies on the specific implication of each isoform in relation to antineoplasic or anti-HIV drug transport across cell membrane would help us to understand drug-drug interactions, drug resistance, and therapy failure and would provide us new insights in relation to drug pharmacokinetics and drug metabolism.

Acknowledgments

We thank Dr. H. Koepsell for helpful discussion and critical manuscript review.

Footnotes

  • This work has been mostly supported by Grants FIPSE 36372/03 and 36621/06, (jointly to M.P.-A. and J.M.-P.). Additional support was provided by Research Grants SAF2005-01259 (to M.P.-A.) and SAF2004-06991, Marató-TV3 021110, and the Spanish AIDS network “Red Temática Cooperativa de Investigación en Sindrome de Immune Deficiencia Adquirida (SIDA) (G03/173 and RD06/006)” (to J.M.-P.). S.P. was supported by Fundación para la Investigación y la Prevención de SIDA en España, and G.M. was supported by Grant 2005FI-00314 from Agència de Gestic d'Ajuts Universitaris i de Recerca-Generalitat de Catalunya.

  • Article, publication date, and citation information can be found at http://jpet.aspetjournals.org.

  • doi:10.1124/jpet.107.131482.

  • ABBREVIATIONS: HIV, human immunodeficiency virus; hCNT, human concentrative nucleoside transporter; hENT, human equilibrative nucleoside transporter; hOAT, human organic anion transporter; hOCT, human organic cation transporter; hOCTN, human organic cation/zwitterion transporter; PHA, phytohemagglutinin; ddC, zalcitabine; ddI, didanosine; IL, interleukin; MDM, monocyte-derived macrophages; D-22, 1,1′-diethyl-2,2′-cyanine iodide; MDDCs, monocyte-derived dendritic cells; NBTI, nitrobenzylthioinosine; DIP, dipyridamole; MPP+, N-methyl-4-phenylpyridinium; ET, ergothioneine; GM-CSF, granulocyte/macrophage colony-stimulating factor; HRP, horseradish peroxidase; AZT, azidothymidine; TBS, Tris-buffered saline; LPS, lipopolysaccharide; PCR, polymerase chain reaction; CT, threshold cycle; mMDDC, mature MDDC; 5′-DFUR, 5′-deoxy-5-fluorouradine; Rani, ranitidine; hNT, human nucleoside transporters; DAB, 3,3′-diaminobenzidine.

    • Received September 10, 2007.
    • Accepted November 26, 2007.
  • The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    Aiba T, Sakurai Y, Tsukada S, and Koizumi T (1995) Effects of probenecid and cimetidine on the renal excretion of 3′-azido-3′-deoxythymidine in rats. J Pharmacol Exp Ther 272: 94–99.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    Aquaro S, Bagnarelli P, Guenci T, De Luca A, Clementi M, Balestra E, Calio R, and Perno CF (2002) Long-term survival and virus production in human primary macrophages infected by human immunodeficiency virus. J Med Virol 68: 479–488.
    OpenUrlCrossRefPubMed
  3. ↵
    Baldwin SA, Beal PR, Yao SY, King AE, Cass CE, and Young JD (2004) The equilibrative nucleoside transporter family, SLC29. Pflugers Arch 447: 735–743.
    OpenUrlCrossRefPubMed
  4. ↵
    Cano-Soldado P, Lorrayoz IM, Molina-Arcas M, Casado FJ, Martinez-Picado J, Lostao MP, and Pastor-Anglada M (2004) Interaction of nucleoside inhibitors of HIV-1 reverse transcriptase with the concentrative nucleoside transporter-1 (SLC28A1). Antivir Ther 9: 993–1002.
    OpenUrlPubMed
  5. ↵
    Chang C, Swaan PW, Ngo LY, Lum PY, Patil SD, and Unadkat JD (2004) Molecular requirements of the human nucleoside transporters hCNT1, hCNT2, and hENT1. Mol Pharmacol 65: 558–570.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Duran JM, Peral MJ, Calonge ML, and Ilundain AA (2005) OCTN3: a Na+-independent L-carnitine transporter in enterocytes basolateral membrane. J Cell Physiol 202: 929–935.
    OpenUrlCrossRefPubMed
  7. ↵
    Errasti-Murugarren E, Pastor-Anglada M, and Casado FJ (2007) Role of Cnt3 in the transepithelial flux of nucleosides and nucleoside-derived drugs. J Physiol 582: 1249–1260.
    OpenUrlCrossRefPubMed
  8. ↵
    Evans WE and Relling MV (1999) Pharmacogenomics: translating functional genomics into rational therapeutics. Science 286: 487–491.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Farre X, Guillen-Gomez E, Sanchez L, Hardisson D, Plaza Y, Lloberas J, Casado FJ, Palacios J, and Pastor-Anglada M (2004) Expression of the nucleoside-derived drug transporters hCNT1, hENT1 and hENT2 in gynecologic tumors. Int J Cancer 112: 959–966.
    OpenUrlCrossRefPubMed
  10. ↵
    Fridland A, Connelly MC, and Robbins BL (2000) Cellular factors for resistance against antiretroviral agents. Antivir Ther 5: 181–185.
    OpenUrlPubMed
  11. ↵
    Gray JH, Owen RP, and Giacomini KM (2004) The concentrative nucleoside transporter family, SLC28. Pflugers Arch 447: 728–734.
    OpenUrlCrossRefPubMed
  12. ↵
    Grundemann D, Harlfinger S, Golz S, Geerts A, Lazar A, Berkels R, Jung N, Rubbert A, and Schomig E (2005) Discovery of the ergothioneine transporter. Proc Natl Acad SciUSA 102: 5256–5261.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Hong M, Schlichter L, and Bendayan R (2001) A novel zidovudine uptake system in microglia. J Pharmacol Exp Ther 296: 141–149.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Hu H, Endres CJ, Chang C, Umapathy NS, Lee EW, Fei YJ, Itagaki S, Swaan PW, Ganapathy V, and Unadkat JD (2006) Electrophysiological characterization and modeling of the structure activity relationship of the human concentrative nucleoside transporter 3 (hCNT3). Mol Pharmacol 69: 1542–1553.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Jutel M, Watanabe T, Akdis M, Blaser K, and Akdis CA (2002) Immune regulation by histamine. Curr Opin Immunol 14: 735–740.
    OpenUrlCrossRefPubMed
  16. ↵
    Kock K, Grube M, Jedlitschky G, Oevermann L, Siegmund W, Ritter CA, and Kroemer HK (2007) Expression of adenosine triphosphate-binding cassette (ABC) drug transporters in peripheral blood cells: relevance for physiology and pharmacotherapy. Clin Pharmacokinet 46: 449–470.
    OpenUrlCrossRefPubMed
  17. ↵
    Koepsell H and Endou H (2004) The SLC22 drug transporter family. Pflugers Arch 447: 666–676.
    OpenUrlCrossRefPubMed
  18. ↵
    Koepsell H, Lips K, and Volk C (2007) Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications. Pharmacol Res 24: 1227–1251.
    OpenUrlCrossRef
  19. ↵
    Lee W and Kim RB (2004) Transporters and renal drug elimination. Annu Rev Pharmacol Toxicol 44: 137–166.
    OpenUrlCrossRefPubMed
  20. ↵
    Leung S and Bendayan R (2001) Uptake properties of lamivudine (3TC) by a continuous renal epithelial cell line. Can J Physiol Pharmacol 79: 59–66.
    OpenUrlCrossRefPubMed
  21. ↵
    Mahony WB, Domin BA, Daluge SM, and Zimmerman TP (2004) Membrane permeation characteristics of abacavir in human erythrocytes and human T-lymphoblastoid CD4+ CEM cells: comparison with (–)-carbovir. Biochem Pharmacol 68: 1797–1805.
    OpenUrlCrossRefPubMed
  22. ↵
    Mangravite LM, Xiao G, and Giacomini KM (2003) Localization of human equilibrative nucleoside transporters, hENT1 and hENT2, in renal epithelial cells. Am J Physiol Renal Physiol 284: F902–F910.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Mata JF, Garcia-Manteiga JM, Lostao MP, Fernandez-Veledo S, Guillen-Gomez E, Larrayoz IM, Lloberas J, Casado FJ, and Pastor-Anglada M (2001) Role of the human concentrative nucleoside transporter (hCNT1) in the cytotoxic action of 5′-deoxy-5-fluorouridine, an active intermediate metabolite of capecitabine, a novel oral anticancer drug. Mol Pharmacol 59: 1542–1548.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    McRae MP, Lowe CM, Tian X, Bourdet DL, Ho RH, Leake BF, Kim RB, Brouwer KL, and Kashuba AD (2006) Ritonavir, saquinavir, and efavirenz, but not nevirapine, inhibit bile acid transport in human and rat hepatocytes. J Pharmacol Exp Ther 318: 1068–1075.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Molina-Arcas M, Bellosillo B, Casado FJ, Montserrat E, Gil J, Colomer D, and Pastor-Anglada M (2003) Fludarabine uptake mechanisms in B-cell chronic lymphocytic leukemia. Blood 101: 2328–2334.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Molina-Arcas M, Marce S, Villamor N, Huber-Ruano I, Casado FJ, Bellosillo B, Montserrat E, Gil J, Colomer D, and Pastor-Anglada M (2005) Equilibrative nucleoside transporter-2 (hENT2) protein expression correlates with ex vivo sensitivity to fludarabine in chronic lymphocytic leukemia (CLL) cells. Leukemia 19: 64–68.
    OpenUrlPubMed
  27. ↵
    Pastor-Anglada M, Cano-Soldado P, Molina-Arcas M, Lostao MP, Larrayoz I, Martinez-Picado J, and Casado FJ (2005) Cell entry and export of nucleoside analogues. Virus Res 107: 151–164.
    OpenUrlCrossRefPubMed
  28. ↵
    Purcet S, Minuesa G, Molina-Arcas M, Erkizia I, Casado FJ, Clotet B, Martinez-Picado J, and Pastor-Anglada M (2006) 3′-Azido-2′,3′-dideoxythymidine (zidovudine) uptake mechanisms in T lymphocytes. Antivir Ther 11: 803–811.
    OpenUrlPubMed
  29. ↵
    Schneider E, Machavoine F, Pleau JM, Bertron AF, Thurmond RL, Ohtsu H, Watanabe T, Schinkel AH, and Dy M (2005) Organic cation transporter 3 modulates murine basophil functions by controlling intracellular histamine levels. J Exp Med 202: 387–393.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Smith CL, Pilarski LM, Egerton ML, and Wiley JS (1989) Nucleoside transport and proliferative rate in human thymocytes and lymphocytes. Blood 74: 2038–2042.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Soler C, Felipe A, Garcia-Manteiga J, Serra M, Guillen-Gomez E, Casado FJ, MacLeod C, Modolell M, Pastor-Anglada M, and Celada A (2003) Interferongamma regulates nucleoside transport systems in macrophages through signal transduction and activator of transduction factor 1 (STAT1)-dependent and -independent signalling pathways. Biochem J 375: 777–783.
    OpenUrlCrossRefPubMed
  32. ↵
    Soler C, Valdes R, Garcia-Manteiga J, Xaus J, Comalada M, Casado FJ, Modolell M, Nicholson B, MacLeod C, Felipe A, et al. (2001) Lipopolysaccharide-induced apoptosis of macrophages determines the up-regulation of concentrative nucleoside transporters Cnt1 and Cnt2 through tumor necrosis factor-alpha-dependent and -independent mechanisms. J Biol Chem 276: 30043–30049.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Tamai I, Ohashi R, Nezu JI, Sai Y, Kobayashi D, Oku A, Shimane M, and Tsuji A (2000) Molecular and functional characterization of organic cation/carnitine transporter family in mice. J Biol Chem 275: 40064–40072.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Tokuhiro S, Yamada R, Chang X, Suzuki A, Kochi Y, Sawada T, Suzuki M, Nagasaki M, Ohtsuki M, Ono M, et al. (2003) An intronic SNP in a RUNX1 binding site of SLC22A4, encoding an organic cation transporter, is associated with rheumatoid arthritis. Nat Genet 35: 341–348.
    OpenUrlCrossRefPubMed
  35. ↵
    Wang L, Giannoudis A, Lane S, Williamson P, Pirmohamed M, and Clark RE (2007) Expression of the uptake drug transporter hOCT1 is an important clinical determinant of the response to imatinib in chronic myeloid leukemia. Clin Pharmacol Ther, in press.
  36. ↵
    White DL, Saunders VA, Dang P, Engler J, Zannettino AC, Cambareri AC, Quinn SR, Manley PW, and Hughes TP (2006) OCT-1-mediated influx is a key determinant of the intracellular uptake of imatinib but not nilotinib (AMN107): reduced OCT-1 activity is the cause of low in vitro sensitivity to imatinib. Blood 108: 697–704.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Wiley JS, Snook MB, and Jamieson GP (1989) Nucleoside transport in acute leukaemia and lymphoma: close relation to proliferative rate. Br J Haematol 71: 203–207.
    OpenUrlPubMed
  38. ↵
    Zhang J, Visser F, King KM, Baldwin SA, Young JD, and Cass CE (2007) The role of nucleoside transporters in cancer chemotherapy with nucleoside drugs. Cancer Metastasis Rev 26: 85–110.
    OpenUrlCrossRefPubMed
  39. ↵
    Zhang L, Gorset W, Washington CB, Blaschke TF, Kroetz DL, and Giacomini KM (2000) Interactions of HIV protease inhibitors with a human organic cation transporter in a mammalian expression system. Drug Metab Dispos 28: 329–334.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Zhang S, Lovejoy KS, Shima JE, Lagpacan LL, Shu Y, Lapuk A, Chen Y, Komori T, Gray JW, Chen X, et al. (2006) Organic cation transporters are determinants of oxaliplatin cytotoxicity. Cancer Res 66: 8847–8857.
    OpenUrlAbstract/FREE Full Text
Back to top

In this issue

Journal of Pharmacology and Experimental Therapeutics: 385 (1)
Journal of Pharmacology and Experimental Therapeutics
Vol. 385, Issue 1
1 Apr 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Journal of Pharmacology and Experimental Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Expression and Functionality of Anti-Human Immunodeficiency Virus and Anticancer Drug Uptake Transporters in Immune Cells
(Your Name) has forwarded a page to you from Journal of Pharmacology and Experimental Therapeutics
(Your Name) thought you would be interested in this article in Journal of Pharmacology and Experimental Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research ArticleMETABOLISM, TRANSPORT, AND PHARMACOGENOMICS

Expression and Functionality of Anti-Human Immunodeficiency Virus and Anticancer Drug Uptake Transporters in Immune Cells

Gerard Minuesa, Sergi Purcet, Itziar Erkizia, Míriam Molina-Arcas, Margarita Bofill, Nuria Izquierdo-Useros, F. Javier Casado, Bonaventura Clotet, Marçal Pastor-Anglada and Javier Martinez-Picado
Journal of Pharmacology and Experimental Therapeutics February 1, 2008, 324 (2) 558-567; DOI: https://doi.org/10.1124/jpet.107.131482

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Research ArticleMETABOLISM, TRANSPORT, AND PHARMACOGENOMICS

Expression and Functionality of Anti-Human Immunodeficiency Virus and Anticancer Drug Uptake Transporters in Immune Cells

Gerard Minuesa, Sergi Purcet, Itziar Erkizia, Míriam Molina-Arcas, Margarita Bofill, Nuria Izquierdo-Useros, F. Javier Casado, Bonaventura Clotet, Marçal Pastor-Anglada and Javier Martinez-Picado
Journal of Pharmacology and Experimental Therapeutics February 1, 2008, 324 (2) 558-567; DOI: https://doi.org/10.1124/jpet.107.131482
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • HDL Mimetic 4F Modulates Aβ Distribution in Brain and Plasma
  • AOX1 Inhibition by Gefitinib, Erlotinib, and Metabolites
  • Catalytic Activity of CYP2C9 Variants
Show more Metabolism, Transport, and Pharmacogenomics

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About JPET
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0103 (Online)

Copyright © 2023 by the American Society for Pharmacology and Experimental Therapeutics