Abstract
In the central nervous system (CNS), brain macrophages and microglia are the primary targets of productive human immunodeficiency virus 1 (HIV-1) infection. Zidovudine (ZDV), a thymidine derivative, has been reported to reduce the progression of the disease and prolong survival in patients with acquired immunodeficiency syndrome (AIDS) and AIDS dementia complex. Although a restricted ZDV distribution has been observed in the CNS, its accumulation in brain parenchyma has not been examined. We have investigated the uptake properties of radiolabeled ZDV by a continuous rat microglia cell line (MLS-9) grown as a monolayer on an impermeable surface. Although the organic cations verapamil, mepiperphenidol, quinidine, cimetidine, andN1-methylnicotinamide moderately inhibited ZDV uptake, the organic cation probes tetraethylammonium and 1-methyl-4-phenylpyridinium were weak inhibitors. ZDV uptake was significantly increased when the proton gradient was outward (pHi 6.3 < pHo 7.4; pHi∼7.1 < pH 8.0), whereas uptake decreased with extracellular acidification (pHi ∼7.1 > pHo 6.0) or in the presence of the Na+/H+ ionophore monensin. ZDV uptake was increased under depolarized membrane conditions (i.e., 138 mM K+ in external medium) and decreased under hyperpolarized conditions (i.e., 2 mM K+ in external medium), implying a membrane potential dependence. These results suggest that although ZDV transport system in microglia has some specificity features of an organic cation transporter, it involves a carrier, distinct from other cloned organic cation transporters, that is novel in its sensitivity to pH and membrane potential. This system may play a significant role in the transport of other weak organic cation substrates and/or metabolites in brain parenchyma.
Human immunodeficiency virus 1 (HIV-1) infection of the brain causes HIV-1 encephalopathy and AIDS dementia complex (ADC), a syndrome characterized by cognitive, motor, and behavioral disturbances (Navia and Price, 1998). Patients with severe ADC usually present microglial activation and multinucleated giant cells, the hallmarks of HIV encephalitis. In the central nervous system (CNS), it appears that only macrophages and microglia harbor the virus particles and the number of proviruses correlates with HIV encephalitis (Navia and Price, 1998).
Microglia are a distinct population of non-neuronal cells that are involved in maintenance of neuronal homeostasis, synaptic plasticity, and repair. These cells play a key role in CNS immune and inflammatory reactions, and are susceptible to diverse insults, ranging from physical damage to infectious agents (Gonzalez-Scarano and Baltuch, 1999). At the two extremes, microglia morphology is 1) spheroid/activated, a state that appears early in development and after brain lesions, and 2) ramified/resting microglia, present in the normal adult brain. Microglial activation, a process that often includes changes in morphology and surface expression of immune-related molecules, has been described in virtually all human neuropathologies, including neurodegenerative diseases, multiple sclerosis, and HIV-1 infection (Gonzalez-Scarano and Baltuch, 1999). The microglia used in our in vitro cell system, are spheroid, proliferating, and capable of producing nitric oxide (Hong et al., 2000; C. A. Colton and L. C. Schlichter, unpublished data).
For the effective treatment of AIDS-ADC or AIDS encephalopathy, anti-HIV drugs need to reach the CNS in significant amounts. ZDV (3′-azido-3′-deoxythymidine, AZT, Retrovir), a thymidine analog, is a potent inhibitor of the in vitro replication and cytopathic effect of HIV. Although limited CNS access has been reported for most antiretroviral drugs, after ZDV administration, substantial improvement was noted in ADC patients (Fischl et al., 1987). The clinical benefits include increased survival, reduced number of opportunistic infections, and partial improvement in neurological defects. However, ZDV treatment is limited by its hematological toxicity (anemia, neutropenia), the development of HIV resistance, and its limited accumulation in certain tissues, including the CNS, which is an important site for HIV replication.
ZDV can enter the CNS by passive diffusion across the blood-brain barrier (BBB) and, more significantly, the blood-cerebrospinal fluid (CSF) barrier (Thomas and Segal, 1997). However, efflux via a probenecid-sensitive transport system at the BBB contributed to the restricted distribution of ZDV in brain tissue (Takasawa et al., 1997b). Low CSF-to-plasma concentration ratios of ZDV after i.v. infusion have been reported in rats (0.15) (Galinsky et al., 1990), rabbits (0.26) (Wong et al., 1993), and humans (0.5) (Blum et al., 1988). Thus, the low steady-state ZDV levels in the CNS depend on both diffusive and active processes.
Because ZDV can act either as an organic anion or cation, consistent with the resonance structures of the azido moiety and pKa value of 9.68 (Henry et al., 1988), in principle, its transport could involve nucleoside transporters, organic anion or organic cation transporters. Interestingly, ZDV interacts with an organic anion transporter at the basolateral membrane (Griffiths et al., 1991) and an organic cation transporter at the brush-border membrane (Griffiths et al., 1992) of renal cortical vesicles. Consistent with such transport, an energy-dependent organic cation carrier is inhibited by ZDV in opossum kidney cells (Chen et al., 1999). Yao et al. (1996) provided direct evidence for Na+-dependent ZDV transport using heterologous expression of the intestinal/kidney nucleoside transporter rCNT1 in Xenopus oocytes. The lipophilicity imparted by the azido group allows nonfacilitated diffusion of ZDV into human erythrocytes, lymphocytes, macrophages, bone marrow progenitor cells, and intestinal epithelial cells.
Our previous study identified a Na+-dependent nucleoside transporter highly sensitive to ZDV in microglia cells (Hong et al., 2000). However, it is not known which, if any, transporters for ZDV exist in microglia, the primary target of HIV infection in the CNS. The goal of this study was to determine the in vitro disposition of ZDV by a brain parenchymal cell line (MLS-9 microglia cells) to characterize its mechanism of transport, and identify potential interactions with other compounds at the carrier site(s).
Materials and Methods
MLS-9 Culture.
Cultures of MLS-9 microglia cells were prepared as previously described (Zhou et al., 1998). Microglia were originally enriched from neopallia of 2- or 3-day-old Wistar rats (Schlichter et al., 1996) to >98% purity, as judged by staining with isolectin B4, OX-42, and ED-1 antibody (Sigma Chemical Co., St. Louis, MO). Colony-stimulating factor 1 was used to induce proliferation and the continuous microglia cell line MLS-9 was established from one of the colonies that arose after several weeks of culturing. These cells also stained with isolectin B4 (100%), OX-42 antibody (98%), and ED-1 antibody (99%), but were negative for the astrocyte marker glial fibrillary acidic protein, and the fibroblast protein fibronectin (Zhou et al., 1998).
Monolayers of MLS-9 cells (passages 25–38) were maintained in either 75-cm2 Falcon plastic tissue culture flasks or 24-well plates (Becton Dickinson, Lincoln Park, NJ) at 37°C in 95% air, 5% CO2. The cells were cultured in minimal essential medium, pH 7.2, supplemented with l-glutamine,d-glucose, 5% fetal bovine serum, 5% horse serum, and 0.5% penicillin/streptomycin suspension (all obtained from Life Technologies, Grand Island, NY), with a change of medium every 2 days. When confluent, cells were passaged using a sodium citrate solution containing 130 mM NaCl, 15 mM sodium citrate, 10 mM glucose, and 10 mM HEPES, pH 7.4. As previously described (Hong et al., 2000), the morphology of MLS-9 cells in confluent monolayers was spheroid, with large cell body structures and very short uropod-like processes.
Uptake of ZDV.
For uptake experiments, MLS-9 cells were seeded on 2-cm2, 24-well plates at 5 × 105 cells/well, and only those grown to a uniform, confluent monolayer (within 4–5 days) were used. The integrity of cell monolayers was evaluated microscopically, before and after the uptake measurements. Most importantly, the morphology and uptake values were the same among the cell passages used.
The uptake of [3H]ZDV (15 Ci/mmol; Moravek Biochemicals, Brea, CA) was measured as previously described (Bendayan et al., 1994). Briefly, MLS-9 cells were rinsed and preincubated for 30 min with 0.5 ml of Earle's balanced saline solution (EBSS), containing: 1.8 mM CaCl2, 5.4 mM KCl, 0.8 mM MgSO4, 138 mM NaCl, 1.0 mM Na2HPO4, 5.5 mMd-glucose, and 20 mM HEPES, with Trizma base added to bring the pH to 7.4. Then, 0.5 ml of this medium containing [3H]ZDV and nonradioactive ZDV was added to the cells at 37°C for fixed time intervals. For uptake specificity studies, cells were preincubated with an inhibitor before adding ZDV. Uptake of radiolabeled probe was terminated by adding 2 ml of ice-cold “stop” solution (0.16 M NaCl). The cells were then solubilized in 1 ml of 1 N NaOH for 30 min, and the lysate transferred to scintillation vials containing 0.5 ml of 2 N HCl. Intracellular accumulation of [3H]ZDV was quantitated using a Beckman liquid scintillation counter (model LS 7000). The sample counts were corrected for variable quench, “zero time” uptake, and background radioactivity in each experiment. The extracellular space, determined using d-[14C]mannitol (51.5 mCi/mmol; NEN Life Science Products, Boston, MA), was negligible (<0.14%); therefore, no correction was applied. The protein concentration (milligrams per milliliter) in each culture plate was determined by the Bradford method, using BSA as the standard and Bio-Rad reagent (Bio-Rad, Mississauga, Ontario, Canada). ZDV uptake values were expressed in nanomoles per milligram of protein per milliliter. For uptake studies at different external pH values, EBSS was buffered with either 0.1 mM Tris, pH 8.0, or 0.1 mM 2-(N-morpholino)ethanesulfonic acid (MES), pH 6.0. To change the intracellular pH, the cells were preincubated for 15 min with 30 mM NH4Cl, before performing the uptake measurements in standard EBSS (pH 7.4). Spectrofluorometry, using a pH indicator dye (see below), showed that exposure to NH4Cl transiently alkalinized the cells (from pH 7.1 to 7.5) and that washing out the external NH4Cl rapidly acidified them (from pH 7.1 to 6.3). This standard procedure for acidifying intracellular pH involves dissociating trapped NH4+ into NH3, which diffuses out, and protons, which are sequestered inside the cells. For all experiments, [3H]ZDV uptake in the presence of a high concentration (3 mM) of verapamil was used to estimate nonspecific uptake. Whenever ethanol was used as the solvent, i.e., with the potassium ionophore valinomycin (1 μM) or the Na+/H+ ionophore monensin (5 μM), solutions contained the same ethanol concentration. There was no significant difference between [3H]ZDV uptake values in the presence (<0.01%) or absence of ethanol; thus, the cells were not adversely affected.
The nucleoside analog drugs ZDV, lamivudine, and abacavir were a gift from Glaxo Wellcome (Research Triangle Park, NC). Didanosine and zalcitabine were generously provided by Bristol-Myers Squibb (Princeton, NJ) and Hoffmann-La Roche (Nutley, NJ), respectively. Unless specified, other chemicals were from Sigma Chemical Co. (St. Louis, MO) and were of the highest purity available.
Metabolism of ZDV.
Intracellular metabolism of [3H]ZDV during the uptake assay was monitored by thin-layer chromatography. MLS-9 cells were incubated for 1, 10, and 30 min with 0.5 mM [3H]ZDV (100 μCi/ml) at 37°C. After the uptake reaction these cell suspensions, including 1 mM solutions of the standards thymine and ZDV, were spotted and chromatographed for 4 h on 250-μm-thick silica gel-coated plates (silica gel 60; Sigma Chemical Co.) that were impregnated with a fluorescent indicator. The solvent was butan-1-ol saturated with water. When the plate was dried, the zones bearing the standards were located under UV light. The RF values corresponding to ZDV, thymine, and ZDV nucleotides (mono-, di-, and triphosphate metabolites) were 0.83, 0.72, and 0, respectively. Each zone was scraped from the plate and analyzed for radioactivity by standard liquid scintillation counting methods.
Intracellular pH Measurements.
Intracellular pH was measured fluorimetrically using the pH-sensitive carboxyfluorescein derivative 2′,7′-bis(carboxyethyl)-5(6′-carboxyfluorescein (BCECF) (Molecular Probes, Eugene, OR). Single-cell measurements were performed with a Zeiss Axiovert 100TV microscope equipped with a Cohu (model 1412) charge-coupled device camera, which is controlled by the operational software Axon Imaging Work Bench, 2.1. The nonfluorescent, membrane-permeant acetoxymethyl ester BCECF-AM enters cells readily and is cleaved by cytosolic esterases to yield the nonpermeant form whose fluorescence is proportional to pH. Confluent monolayers of MLS-9 cells grown in 24-well tissue culture plates were preincubated (40 min, 37°C) in EBBS containing 20 μM BCECF-AM and 0.1% BSA. Cells were washed and preincubated in 30 mM NH4Cl solution for 30 min, and then followed by its removal and replacement with either EBSS or 1 mM amiloride. Baseline measurements were recorded in EBSS. Fluorescence was detected with dual excitation at 440 and 495 nm, and emission at 535 nm. Emission ratios for 440- versus 495-nm excitation were computed by the software. Intracellular pH was calibrated by comparing the fluorescence intensity of cells with 100 μM BCECF acid in EBSS solution, in which the pH was buffered with 0.1 mM MES (pH 6.1), BES (pH 6.7 and 7.1), HEPES (pH 7.4 and 7.7), and Tris (pH 8.1 and 8.6). All buffers were from Sigma Chemical Co.
Data Analysis.
Experiments were repeated at least two times using cells from different passages. Data points in each experiment represent quadruplicate measurements. Results are presented as mean ± S.D. from a minimum of two separate experiments. The Michaelis-Menten kinetic parameters (Km andVmax) for ZDV transport were determined by a nonlinear least-squares analysis in Sigma Plot 4.0. To estimate the inhibitory constant (Ki) of the inhibitor verapamil, a least-squares regression analysis was used to determine the linear correlation between V andV/[ZDV], the Eadie-Hofstee linear transformation. The IC50 or concentration of various organic cationic drugs causing a 50% reduction in ZDV uptake was estimated by a sigmoidal, four-parameter inhibition model. Statistical significance was assessed by the Student's t test for unpaired experimental values, the test of repeated measures of ANOVA, and/or the post hoc multiple-comparison Bonferroni t test, as appropriate. A p value <0.05 was considered significant.
Results
Metabolism of ZDV.
The intracellular metabolism of ZDV has been examined extensively in cell lines, in retrovirus-infected mouse tissues, and in human peripheral blood mononuclear cells (Furman et al., 1986). To exert its antiviral activity, ZDV is converted by several intracellular kinases to its phosphorylated anabolites. That is, thymidine kinase, thymidylate kinase, and pyrimidine nucleoside diphosphate kinase consecutively convert ZDV to ZDV-monophosphate, ZDV-diphosphate, and ZDV-triphosphate, respectively. Because metabolism can complicate the interpretation of transport studies, we determined the extent of ZDV metabolism by microglia cells. After the uptake reaction, chromatographic analysis of the cell indicated no significant ZDV metabolism before 30 min. For instance, in cells incubated with 0.5 mM [3H]ZDV for 30 min at 37°C, 87% of the intracellular radioactivity was associated with the unmodified form ZDV, 5% cochromatographed with thymine, and 8% with the ZDV nucleotides (mono-, di-, and triphosphate metabolites). After a 10-min incubation, 94, 4, and 2% of the radioactivity was recovered in the ZDV, thymine, and ZDV nucleotide fractions, respectively. Similarly, after a 1-min incubation, 96, 3, and 1% of the radioactivity comigrated with ZDV, thymine, and ZDV nucleotides, respectively (data not shown).
Time Course of Specific ZDV Uptake.
Figure1 shows that specific ZDV (0.5 mM) uptake by MLS-9 cells at 37°C was essentially linear for the first 20 min and began to approach an equilibrium value by ∼30 min. Therefore, a 10-min incubation time was used to represent the initial rate of ZDV influx into the cells. The ZDV concentration (0.5 mM) was chosen after considering the kinetics of the system involved (see below), to approximate the apparent Km value and better describe ZDV uptake properties.
Kinetics of ZDV Uptake.
The initial rate of ZDV influx into MLS-9 cells was measured at 10 min (37°C) with ZDV concentrations from 20 μM to 2.5 mM. The total ZDV uptake rate (V) can be described by the following equation:
Specificity Studies.
ZDV has previously been shown to inhibit an organic cation transporter in rat renal brush-border membrane vesicles (Griffiths et al., 1992). Thus, to determine the selectivity of the transporter, we tested several organic cations (1 mM) for their ability to inhibit the influx of 0.5 mM [3H]ZDV, measured at 10 min and 37°C (Fig.3). Among these compounds, verapamil, mepiperphenidol, quinidine, cimetidine, andN1-methylnicotinamide, known inhibitors of organic cation transport systems, were the most effective inhibitors (70–79% inhibition). Thiamine, 1-methyl-4-phenylpyridinium (MPP+), guanidine, tetraethylammonium (TEA), and trimethoprim (49–67% inhibition) were weak inhibitors. Because these results suggested the possible involvement of an organic cation transport (OCT) system for ZDV, we also explored the effect of ZDV on the uptake of radiolabeled TEA, a standard organic cation probe. As opposed to findings with all the other cloned OCT systems (i.e., OCT1, OCT2, OCT3, OCTN1, and OCTN2), which are readily known to transport TEA (Gorboulev et al., 1997; Kekuda et al., 1998; Wu et al., 1998b,c), the uptake of TEA by microglia cells was low and not sensitive to ZDV (data not shown).
Further inhibitory studies were undertaken and IC50 values for the moderate inhibitors were determined by fitting the data to a sigmoidal equation:
We next explored the type of inhibition exerted by the most potent inhibitor we identified. Verapamil inhibited ZDV uptake in a competitive manner with an estimated inhibitory constant (Ki) of 167 ± 21 μM. TheKi value was obtained from the competitive inhibition equation:
Because previous studies have demonstrated that ZDV is both a substrate for and competitive inhibitor of the organic anion transporter in rat renal basolateral membrane vesicles (Griffiths et al., 1991), we explored the inhibitory effect of several organic anions on ZDV uptake by MLS-9 cells. Organic anions such as benzyl penicillin, salicylic acid, and the prototypic substrate p-aminohippuric acid had no significant effect on ZDV uptake (data not shown). Moreover, contrary to the ZDV efflux systems identified at the blood-brain barrier and blood-CSF barrier (Takasawa et al., 1997a,b), ZDV transporter in microglia was insensitive to probenecid, a known inhibitor of organic anion transporters (data not shown).
Nucleoside transporters (i.e., rCNT1) have also been shown to mediate ZDV transport (Yao et al., 1996). Thus, we explored the involvement of a nucleoside transport system (either equilibrative or concentrative) by testing the inhibitory effect of nucleosides (thymidine, cytidine, guanosine, and adenosine), nucleoside analogs (lamivudine, abacavir, didanosine, and zalcitabine), and standard nucleoside transport inhibitors [dilazep, dipyridamole, and 6-(4-nitrobenzyl)-thio-9-β-d-ribofuranosylpurine] on ZDV uptake, in the presence and absence of Na+(Na+ replaced withN-methyl-d-glucamine). No significant difference (P > 0.9) was observed between the control and any of the tested conditions (data not shown), suggesting that ZDV entry into the microglia cells does not involve a nucleoside transporter. These results corroborate our previous finding that ZDV acts only as a potent, noncompetitive inhibitor of the Na+-dependent nucleoside transporter (Hong et al., 2000).
pH and Membrane Potential Effects.
Several studies have identified organic cation/proton exchange mechanisms in brush-border membrane vesicles from kidney and intestine, and in the basolateral membranes of hepatocytes (Zhang et al., 1998). The driving force, a proton gradient, is known to be functionally linked to both the Na+/H+ antiporter and the H+-ATPase (Bendayan et al., 1994). To assess the presence of such a mechanism, we measured ZDV uptake in the presence of an outwardly directed proton gradient, generated by a standard NH4Cl acidification procedure (Jans et al., 1987;Bendayan et al., 1994). Acidification was confirmed by monitoring the changes in intracellular pH with the fluorescent probe BCECF (Fig.5). Addition of NH4Cl caused an initial intracellular alkalinization (to pH 7.5), followed by a dramatic acidification upon NH4Cl removal (from pH 7.1 to 6.3). The pH spontaneously recovered to its initial value by ∼20 min. After a second NH4Cl treatment and washout, the pH recovery was abolished by 1 mM amiloride, an inhibitor of the Na+/H+ antiporter. ZDV uptake was enhanced for the first 5 min, and then decreased to about control values by 10 min, presumably due to the dissipation of the proton gradient (Fig. 6). The converse effect of a proton gradient in the opposite direction (Fig.7) further supports a mechanism whereby protons exchange with ZDV in microglia cells. That is, the initial uptake value was increased by extracellular alkalinization (pHi ∼ 7.1 < pHo8.0), and decreased by extracellular acidification (pHi ∼ 7.1 > pH06.0). Thus, ZDV uptake appears to be increased by an outwardly directed proton gradient and decreased by an inwardly directed one. The requirement for an outwardly directed proton gradient was further supported by the decrease in uptake (by 35%) after exposure to 5 μM monensin (Fig. 8), a Na+/H+ ionophore that collapses the pH gradient. Taken together, these results suggest that proton efflux is required to drive the entry of ZDV into MLS-9 cells.
MLS-9 cells express a K+ current that presumably sets the cell membrane potential (Zhou et al., 1998) in both NaCl and KCl solutions. To determine whether ZDV uptake is electrogenic, membrane potential was changed by varying extracellular K+ concentrations (NaCl replaced by equimolar KCl) to depolarized (138 mM K+), hyperpolarized (2 mM K+), and control (5 mM K+) conditions. With 138 mM K+ in the uptake buffer, ZDV uptake increased by 50%, when measured after 10 min (Fig.9). Because no further increase in uptake was observed when 1 μM valinomycin, a potassium ionophore, was added to the KCl solution, the cells were effectively depolarized by the high concentration of K+. Conversely, when the cells were hyperpolarized with valinomycin and reduced external K+, ZDV uptake was significantly reduced (Fig.9). These results are consistent with a transporter that is membrane potential-dependent.
Discussion
ZDV, a thymidine analog, is considered one of the first-line therapeutic agents for AIDS and AIDS dementia complex (Fischl et al., 1987). To be active, intracellular ZDV must be phosphorylated to its triphosphate derivative and incorporated into the viral DNA by HIV reverse transcriptase, resulting in the termination of DNA elongation (Furman et al., 1986). At 1 to 5 μM concentrations, ZDV is an effective and selective inhibitor of viral DNA replication in vitro.
Penetration of anti-HIV drugs into the CNS is of clinical concern due to the neurological effects of HIV (Navia and Price, 1998). Results from in vivo studies examining ZDV uptake into brain, after a single pass through the cerebral circulation, have demonstrated nonsignificant net transport of ZDV across the BBB (Terasaki and Pardridge, 1988; Wu et al., 1998a). Moreover, Thomas and Segal (1997) have determined, using a ventriculocisternal perfusion technique, that ZDV influx across the blood-CSF and BBB occurs primarily by a diffusional process. Because at present no information is available on the mechanism of transport of nucleoside analog drugs within brain parenchyma, the purpose of this study was to characterize the properties of ZDV uptake by microglia, the major target of HIV infection in the brain.
Kinetic analysis of ZDV uptake by microglia shows a saturable, low-affinity system (Km = 1 mM). Similarly, weak interactions have been reported for ZDV in rat renal membrane vesicles, where ZDV inhibited transport of the organic cationN1-methylnicotinamide at the brush-border membrane and transport of the organic anionp-aminohippuric acid at the basolateral site, with IC50 values of 2.5 mM and 225 μM, respectively (Griffiths et al., 1991, 1992). These in vitro experiments further support in vivo results that showed that in the kidney, ZDV is transported by a basolateral probenecid-sensitive organic anion transporter, and an apical cimetidine-sensitive organic cation transporter (Aiba et al., 1995). Interestingly, a highKm (0.5 mM) has been reported in the transport of ZDV by a recombinant Na+-dependent nucleoside transporter, functionally expressed in Xenopusoocytes (Yao et al., 1996).
Specific transporters responsible for handling cationic compounds have been reported mainly in the kidney, liver, and intestine. Three polyspecific, potential-sensitive organic cation transporters (OCT1, rOCT2/hOCT2, OCT3) cloned originally from rat kidney, and placenta, are differentially expressed in brain (Zhang et al., 1998). Sensitive reverse transcription-polymerase chain reaction and Northern blot studies reported low brain expression of OCT1 and OCT2, although several cationic neurotoxins and neurotransmitters are accepted as substrates by both transporters (Gorboulev et al., 1997; Grundemann et al., 1997). In contrast, mRNA transcripts specific for OCT3 were detected in significant amounts by Northern analysis in brain (i.e., cerebral cortex, cerebellum, hippocampus) and several other tissues (Kekuda et al., 1998; Wu et al., 1998b). Not only is OCT3 capable of transporting various cationic neurotoxins (i.e., MPP+) and neurotransmitters but also it has been shown to exhibit transport properties of an extraneuronal monoamine transporter (Wu et al., 1998b). Recently, two other cloned, electroneutral, H+-driven organic cation transporters, OCTN1 and OCTN2, were shown to be expressed widely in human tissues, including the brain (Tamai et al., 1997; Wu et al., 1998c).
Results from our substrate specificity studies show that several endogenous and exogenous organic cations (i.e.,N1-methylnicotinamide, quinidine, mepiperphenidol, verapamil) inhibit specific ZDV uptake in a concentration-dependent manner with verapamil being a competitive inhibitor. However, the inhibition is modest (IC50 range = 156–200 μM) and the system was weakly inhibited by the OCT probes TEA and MPP+. Furthermore, we observed that the cell line was unable to efficiently transport TEA. This specificity pattern is not shown for other OCT members such as OCT1, OCT2, OCT3, OCTN1, and OCTN2, which are all known to readily transport TEA (Gorboulev et al., 1997; Grundemann et al., 1997; Kekuda et al., 1998; Wu et al., 1998b,c). Thus, despite some OCT specificity similarities, the characterized transporter for ZDV in microglia appears to be distinct from any of the other cloned members of the OCT family.
Several studies have characterized the involvement of a H+/organic cation transporter in the renal luminal transport of organic cations. The driving force, a proton gradient, is known to be functionally linked to both the Na+/H+ antiporter and the H+-ATPase (Bendayan et al., 1994). To elucidate the energetics in ZDV transport by microglia, we investigated the effect of an outwardly directed proton gradient generated by a standard NH4Cl loading procedure (Jans et al., 1987;Bendayan et al., 1994; Faff et al., 1996). The 20-min recovery of internal pH to baseline 7.1 from an intracellular acidification (pH 6.3, a decrease of 0.8 units) was confirmed by microfluorimetry. Moreover, the diuretic drug amiloride, a known inhibitor of the Na+/H+ antiporter, abolished the pH recovery and maintained the cell acidification, suggesting functional activity of a Na+/H+ antiporter. It is well established that amiloride rapidly and competitively inhibits the Na+ site of the Na+/H+ antiporter without affecting other Na+-coupled transport processes, shown, for instance, by its selective inhibition of the Na+/H+ exchanger in rabbit renal microvillus membrane vesicles (Kinsella and Aronson, 1981). Our studies show that with intracellular acidification, ZDV uptake is significantly enhanced within the first 5 min. The accumulation of ZDV cannot be explained by nonionic diffusion because the zwitterionic nature of the molecule is not modified within the pH range studied (6.3–8.0). Furthermore, ZDV uptake was significantly reduced (i.e., by 35% at 30 min) in the presence of the Na+/H+ ionophore monensin, thus suggesting the involvement of a H+exchanger. In mouse microglia, several pH regulatory systems (i.e., Na+/HCO3−cotransporter, Cl−/HCO3−exchanger, H+/K+- ATPase, Na+/H+ antiporter) are functionally expressed (Faff et al., 1996; Shirihai et al., 1998).
To test for membrane potential effects, ZDV uptake was investigated in high- and low-K+ medium in the presence of the potassium ionophore valinomycin. Membrane depolarization enhanced ZDV uptake, whereas hyperpolarization caused a decrease in ZDV uptake. Unlike other potential-driven OCT systems (Kekuda et al., 1998; Wu et al., 1998b), when depolarized with high extracellular K+, an inside positive potential difference drives an increased uptake of ZDV in microglia cells. Thus, although ZDV uptake system in microglia has some specificity features of an organic cation transporter, our results suggest a system that is novel in its sensitivity to membrane potential and pH. Various normal mechanisms that depolarize microglia could enhance ZDV uptake and, ultimately, increase its pharmacological activity. Activation of a number of channels has been reported to depolarize microglia, i.e., Ca2+ (Colton et al., 1994), Na+ (Korotzer and Cotman, 1992), anion channels (Schlichter et al., 1996), or purinergic receptor-gated channels (Illes et al., 1996).
In our cell system, the prototypical organic anionp-aminohippuric acid and the inhibitor probenecid had no effect on ZDV uptake, implying the lack of involvement of an organic anion transporter in MLS-9 cells. In addition, ZDV accumulation by MLS-9 cells was insensitive to a number of nucleosides, nucleoside analog drugs, and standard nucleoside transport inhibitors, suggesting that nucleoside transporters are not involved. These findings are consistent with reports documenting that the absence of the 3′-hydroxy group of the ribose moiety greatly reduces the ability of compounds to be transported by equilibrative nucleoside transporters. Furthermore, ZDV, 2′,3′-dideoxyadenosine, and 2′,3′-dideoxycytidine (zalcitabine) did not have measurable affinity for the BBB nucleoside transporter (Terasaki and Pardridge, 1988) or the Na+-dependent nucleoside transporter at the choroid plexus (Wu et al., 1994). In microglia, we have previously observed that ZDV was a potent noncompetitive inhibitor of a Na+-nucleoside transporter (Hong et al., 2000).
In summary, a novel, electrogenic, proton-driven ZDV transporter has been characterized in microglia, the target and reservoir of HIV infection in the brain. This system may play a significant role in the transport of other weak organic cation substrates and metabolites in brain parenchyma. The low affinity of ZDV for the rat microglia transporter suggests that drug-drug interactions may readily occur at the transporter site(s) with other weak organic cations, most likely causing either a decrease in therapeutic efficacy, an increase of toxicity, or both in AIDS patients receiving routinely multiple drug regimens. Moreover, because interspecies differences in the kinetic and selectivity properties of organic cation transporter homologs from rodent, rabbit, and human have been reported (Dresser et al., 2000), it is possible that a ZDV transporter with different kinetics may be expressed in humans. This could result in substantial differences in the in vivo microglial handling of drugs (i.e., higher affinity transport system for ZDV), basic metabolites, and toxins. It remains to be established whether other physiological roles can be attributed to this transport system.
Acknowledgment
We thank Dr. Peter Pennefather for very helpful comments in the preparation of the manuscript.
Footnotes
-
Send reprint requests to: Dr. Reina Bendayan, Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, 19 Russell St., Toronto, Ontario M5S 2S2, Canada. E-mail:r.bendayan{at}utoronto.ca
-
↵1 Current address: Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 2S2, Canada.
-
↵2 Current address: Cellular and Molecular Biology, Toronto Western Research Institute, University Health Network, Toronto, Ontario M5T 2S8 and Department of Physiology and Institute for Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A1, Canada.
-
This work was supported by a grant from the Ontario HIV Treatment Network (OHTN), the Canadian Foundation for AIDS Research (CANFAR), the Glaxo Wellcome Positive Action Fund, Ontario Ministry of Health, and the Heart and Stroke Foundation of Ontario (no. T3726). M.H. is a recipient of an Ontario HIV Treatment Network Studentship Award.
-
This work was presented in preliminary form (Hong et al., 1999). Poster presented at the 1999 Annual Meeting of the American Association of Pharmaceutical Scientists, New Orleans, LA, Nov. 1999.
- Abbreviations:
- HIV-1
- human immunodeficiency virus 1
- AIDS
- acquired immunodeficiency syndrome
- ADC
- AIDS dementia complex
- CNS
- central nervous system
- ZDV
- zidovudine
- BBB
- blood-brain barrier
- CSF
- cerebrospinal fluid
- EBSS
- Earle's balanced saline solution
- MES
- 2-(N-morpholino)ethanesulfonic acid
- BCECF
- 2′,7′-bis(carboxyethyl)-5(6′-carboxyfluorescein
- BCECF-AM
- 2′,7′-bis(carboxyethyl)-5(6′-carboxyfluorescein acetoxymethyl ester
- MPP+
- 1-methyl-4-phenylpyridinium
- TEA
- tetraethylammonium
- OCT
- organic cation transport
- BES
- N,N-bis(2-hydroxyethyl]-2-aminoethanesulfonic acid
- Received May 19, 2000.
- Accepted September 19, 2000.
- The American Society for Pharmacology and Experimental Therapeutics