characterisation of the endogenous human peripheral...

10
Characterisation of the endogenous human peripheral serotonin transporter SLC6A4 reveals surface expression without N-glycosylation Anita Chamba a, , Michelle J. Holder a , Nicholas M. Barnes b , John Gordon a a Division of Immunity and Infection, The Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdom b Division of Neuroscience, The Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdom abstract article info Article history: Received 20 March 2008 Received in revised form 10 July 2008 Accepted 16 July 2008 Keywords: Serotonin transporter Lymphocytes Glycosylation RT-PCR conrmed that human cell lines of diverse peripheral origins express transcripts for the serotonin transporter (sert/slc6a4). Molecular weights reported for the translated protein appear to be quite variable however. Here we compared directly immunoreactive protein generated from cloned sert transfected into HEK293 with that carried endogenously among the cell lines. The dominant glycosylated 8595 kDa immunoreactive species contained in HEK-sert transfectants was poorly represented in any native cell: instead, discrete 70 and 60 kDa bands were universally detected. Biotinylation of lymphoid cells revealed that the endogenous non-glycosylated 60 kDa but not 70 kDa protein was available at the surface to access exogenous ligands. © 2008 Elsevier B.V. All rights reserved. 1. Introduction The neuronal serotonin transporter (SERT; SLC6A4) has received considerable attention in recent years. As well as its crucial role in the process of neurotransmitter recycling, it represents an established target for many pharmacological agents that affect brain function including antidepressants and psychostimulants. Serotonergic signal- ling is initiated by the release of serotonin (5-HT) from the pre- synaptic neuronal cell into the synaptic cleft. Termination of neuro- transmitter action, a key step that determines the intensity and duration of signalling, occurs by re-uptake of the extracellular serotonin back into nerve terminals through SERT, a high afnity plasma membrane transporter. SERT belongs to the SLC6 gene family of related neurotransmitter proteins, which includes the transporters for dopamine (DAT), norepinephrine (NET), γ-aminobutyric acid (GAT) and glycine (GLYT1 and GLYT2) (Torres et al., 2003). Deregula- tions in serotonergic activity have been implicated in the aetiology of psychiatric diseases such as depression in addition to a large group of compulsive disorders (Malison et al., 1998; Purselle and Nemeroff, 2003). Consequently, there has been much focus on multiple compounds which inhibit SERT function and hence increase the availability of 5-HT: these include tricyclic antidepressants and the selective serotonin re-uptake inhibitors (SSRIs) (Marcusson and Ross, 1990; Ramamoorthy et al., 1993). SERT is also targeted by the drugs of abuse, cocaine (Rothman and Baumann, 2003) and 3,4-methylene- dioxymethamphetamine (ecstasy)(Rudnick and Wall, 1992). Although early SERT studies largely focussed on the expression of the transporter in brain tissue, it is becoming increasingly appar- ent that SERT also exists in specialised non-neuronal cells in the periphery, including platelets (Launay et al., 1992), lymphocytes (Faraj et al., 1994), placental syncytiotrophoblasts (Balkovetz et al., 1989) and epithelial cells of the gastrointestinal mucosa (Wade et al., 1996). Our own interest in human B lymphocytes has revealed functional responses to 5-HT and expression of SERT in the Burkitt's lymphoma cell lines (Serafeim et al., 2002) and in a wider spectrum of B-cell malignancy (Meredith et al., 2005b). The exact function or signicance of SERT in lymphocytes is unknown but a recent review by our group presented evidence for these and other immune cells expressing multiple components of the serotonergic and related dopaminergic systems (Gordon and Barnes, 2003). The increased interest in the determination of both the molecular characteristics and regulation of SERT has subsequently led to the production of a number of specic antibodies. A review of the literature (Table 1) indicates wide discrepancies and uncertainty in reported molecular sizes for SERT, with diverse patterns and characteristics of SERT expression emer- ging. These studies include SERT generated from heterologous expression in cell lines (Qian et al., 1995; Ramamoorthy and Blakely, 1999; Ramamoorthy et al., 1998) others on endogenous SERT expressed constitutively in brain or platelets; and certain other peripheral tissues: for example, lymphoid (Tsao et al., 2006), intestinal mucosa (Bian et al., 2007) and arterial smooth muscle (Ni et al., 2004). Differences in SERT mobility have been attributed to post-transla- tional modications of the protein and there is certainly increasing evidence from mutational studies (Tate and Blakely, 1994) and experiments on related transporters DAT and NET (Cai et al., 2005; Melikian et al., 1996), that N-linked glycosylation may be an important feature for the correct folding of the transporter protein to a fully Journal of Neuroimmunology 204 (2008) 7584 Corresponding author. Tel.: +44 121 414 6970; fax: +44 121 414 3454. E-mail address: [email protected] (A. Chamba). 0165-5728/$ see front matter © 2008 Elsevier B.V. All rights reserved. doi:10.1016/j.jneuroim.2008.07.014 Contents lists available at ScienceDirect Journal of Neuroimmunology journal homepage: www.elsevier.com/locate/jneuroim

Upload: others

Post on 27-Apr-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

Characterisation of the endogenous human peripheral serotonin transporter SLC6A4reveals surface expression without N-glycosylation

Anita Chamba a,⁎, Michelle J. Holder a, Nicholas M. Barnes b, John Gordon a

a Division of Immunity and Infection, The Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdomb Division of Neuroscience, The Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdom

a b s t r a c ta r t i c l e i n f o

Article history:Received 20 March 2008Received in revised form 10 July 2008Accepted 16 July 2008

Keywords:Serotonin transporterLymphocytesGlycosylation

RT-PCR confirmed that human cell lines of diverse peripheral origins express transcripts for the serotonintransporter (sert/slc6a4). Molecular weights reported for the translated protein appear to be quite variablehowever. Here we compared directly immunoreactive protein generated from cloned sert transfected intoHEK293 with that carried endogenously among the cell lines. The dominant glycosylated 85–95 kDaimmunoreactive species contained in HEK-sert transfectants was poorly represented in any native cell:instead, discrete 70 and 60 kDa bands were universally detected. Biotinylation of lymphoid cells revealed thatthe endogenous non-glycosylated 60 kDa but not 70 kDa protein was available at the surface to accessexogenous ligands.

© 2008 Elsevier B.V. All rights reserved.

1. Introduction

The neuronal serotonin transporter (SERT; SLC6A4) has receivedconsiderable attention in recent years. As well as its crucial role in theprocess of neurotransmitter recycling, it represents an establishedtarget for many pharmacological agents that affect brain functionincluding antidepressants and psychostimulants. Serotonergic signal-ling is initiated by the release of serotonin (5-HT) from the pre-synaptic neuronal cell into the synaptic cleft. Termination of neuro-transmitter action, a key step that determines the intensity andduration of signalling, occurs by re-uptake of the extracellularserotonin back into nerve terminals through SERT, a high affinityplasma membrane transporter. SERT belongs to the SLC6 gene familyof related neurotransmitter proteins, which includes the transportersfor dopamine (DAT), norepinephrine (NET), γ-aminobutyric acid(GAT) and glycine (GLYT1 and GLYT2) (Torres et al., 2003). Deregula-tions in serotonergic activity have been implicated in the aetiology ofpsychiatric diseases such as depression in addition to a large group ofcompulsive disorders (Malison et al., 1998; Purselle and Nemeroff,2003). Consequently, there has been much focus on multiplecompounds which inhibit SERT function and hence increase theavailability of 5-HT: these include tricyclic antidepressants and theselective serotonin re-uptake inhibitors (SSRIs) (Marcusson and Ross,1990; Ramamoorthy et al., 1993). SERT is also targeted by the drugs ofabuse, cocaine (Rothman and Baumann, 2003) and 3,4-methylene-dioxymethamphetamine (“ecstasy”) (Rudnick and Wall, 1992).

Although early SERT studies largely focussed on the expressionof the transporter in brain tissue, it is becoming increasingly appar-ent that SERT also exists in specialised non-neuronal cells in theperiphery, including platelets (Launay et al., 1992), lymphocytes (Farajet al., 1994), placental syncytiotrophoblasts (Balkovetz et al., 1989) andepithelial cells of the gastrointestinal mucosa (Wade et al., 1996). Ourown interest in human B lymphocytes has revealed functionalresponses to 5-HT and expression of SERT in the Burkitt's lymphomacell lines (Serafeim et al., 2002) and in a wider spectrum of B-cellmalignancy (Meredith et al., 2005b). The exact function or significanceof SERT in lymphocytes is unknown but a recent review by our grouppresented evidence for these and other immune cells expressingmultiple components of the serotonergic and related dopaminergicsystems (Gordon and Barnes, 2003). The increased interest in thedetermination of both the molecular characteristics and regulationof SERT has subsequently led to the production of a number of specificantibodies. A review of the literature (Table 1) indicates widediscrepancies and uncertainty in reported molecular sizes for SERT,with diverse patterns and characteristics of SERT expression emer-ging. These studies include SERT generated from heterologousexpression in cell lines (Qian et al., 1995; Ramamoorthy and Blakely,1999; Ramamoorthy et al., 1998) others on endogenous SERTexpressed constitutively in brain or platelets; and certain otherperipheral tissues: for example, lymphoid (Tsao et al., 2006), intestinalmucosa (Bian et al., 2007) and arterial smooth muscle (Ni et al., 2004).Differences in SERT mobility have been attributed to post-transla-tional modifications of the protein and there is certainly increasingevidence from mutational studies (Tate and Blakely, 1994) andexperiments on related transporters DAT and NET (Cai et al., 2005;Melikian et al., 1996), that N-linked glycosylationmay be an importantfeature for the correct folding of the transporter protein to a fully

Journal of Neuroimmunology 204 (2008) 75–84

⁎ Corresponding author. Tel.: +44 121 414 6970; fax: +44 121 414 3454.E-mail address: [email protected] (A. Chamba).

0165-5728/$ – see front matter © 2008 Elsevier B.V. All rights reserved.doi:10.1016/j.jneuroim.2008.07.014

Contents lists available at ScienceDirect

Journal of Neuroimmunology

j ourna l homepage: www.e lsev ie r.com/ locate / jneuro im

Page 2: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

active form and therefore contribute to the regulation of traffickingand surface expression of SERT, DAT and NET.

In the present study we have used distinct antibodies directedto sequences at or close to the respective terminal regions (aa51–66and aa611–630) to characterise SERT in human cell lines of diverseperipheral origins, comparing endogenous protein to that generatedby heterologous (over)expression of cDNA encoding full-lengthhuman SERT. The importance of N-glycosylation to generatingdifferent SERT species was examined using PNGase F and tunicamycin.Biotinylation of cell surface proteins and confocal microscopy allowedthe exploration of which endogenous SERT forms reached and wereaccessible at the outer membrane. Our results help clarify some of thediscrepancy inherent in the literature with regards reported char-acteristics of SERT protein and demonstrate that a non-glycosylatedconstitutive 60 kDa form found endogenously (as opposed to beinggenerated via heterologous expression in transfected cells), is capableof trafficking to the outer membrane of peripheral cells and is thedominant species found there. Such information may assist drugdesign where surface SERT offers itself as a potential therapeutictarget in the periphery: as in the case of B-cell malignancy (Meredithet al., 2005b; Serafeim et al., 2002).

2. Materials and methods

2.1. Cells

Stable Bcl-2 transfectants of EBV-ve L3055 Burkitt's lymphoma cells(L3/Bcl-2) weremaintained in RPMI 1640medium (Invitrogen, Paisley,Scotland) supplemented with 10% Serum Supreme (BioWhittaker,Wokingham, U.K.), 2 mM glutamine, 100 IU/ml penicillin and 100 µg/ml streptomycin (Invitrogen) as previously described (Serafeim et al.,2002). These cells are substantially more robust than the parentalwild-type L3055 cells making them amenable to manipulations suchas in the biotinylation experiments (below). A stable line of HEK293cells carrying full-length human SERT (HEK-hSERT) (Qian et al., 1997)and parental HEK293 were a generous gift from Randy Blakely(Vanderbilt University, Nashville, TN) and maintained in monolayerculture in DMEM medium (Invitrogen) containing 10% FCS (First link,Birmingham, U.K.) and selected with G418 sulphate (250 µg/ml).Burkitt's lymphoma Daudi and B-lymphoblastoid cell line RPMI8866were cultured in RPMI 1640 as described for L3/Bcl-2. NeuroblastomaKelly and Jurkat T lymphocytes were maintained in RPMI 1640containing 10% FCS, 2 mM glutamine, 100 IU penicillin and 100 µg/ml

Table 1Comparison of reported SERT sizes

Species Cell Source Size (kDa)⁎ Reference Antibody†

Human HEK293 cDNA 96, 76 (Ramamoorthy et al., 1998) CT2Human Fibroblast cDNA 86, 65 (Wersinger et al., 2006) C-20Human HEK293 cDNA 85–95, 60 (Frankhauser et al., 2006) C-20Human Platelet Endogenous 100, 90Human Platelet Endogenous 68 (Launay et al., 1992) (Coomassie stain)Human Platelet Endogenous 105–68, 43 (Belous et al., 2001) CTHuman Platelet Endogenous 80 (Little et al., 2006) AB1594§

Human Lymphoid Endogenous 80, 65 (Tsao et al., 2006) AB1594Human Brain Endogenous 67 (Rotondo et al., 1996) (Silver stain)Human Platelet Endogenous 70 (Pizzinat et al., 1999) SC (goat)Rat Mesangial Endogenous 70Monkey Brain Endogenous 78 (Shively et al., 2003) ST51-2Human Caco-2 Endogenous 70 (Iceta et al., 2006) AB1594Rat Intestinal mucosa Endogenous 70, 40Rat Brain Endogenous 70, 40Human Lymphocytes Endogenous 100 (Barkan et al., 2004) SC-13997Rat Brain Endogenous 64 (561–630)Rat HeLa cDNA 61 (Qian et al., 1995) S365 CTRat Platelet Endogenous 94 (60?) (388–401)Rat Brain Endogenous 76Rat Brain Endogenous 92, 74, 64 (Zhou et al., 1996) 5-HTT (55–68)

74, 64 5-HTT (315–325)Rat Brain Endogenous 70, 50 (Wang et al., 2005) NT (1–15)Rat Brain Endogenous 76 (Najib et al., 2000) AB1594Rat Brain Endogenous 70 (Rothman et al., 2003) AB1594Rat Brain Endogenous 70‡, 63–68 (Xie et al., 2006) CT/(579–599)Rat Platelet Endogenous 100 (Jayanthi et al., 2005) CT (596–630)Rat Platelet Endogenous 94 (Hranilovic et al., 2001) CTRat Arterial Endogenous 74 (Ni et al., 2004) CT H115 (516–630)Rat HEK293 cDNA 180–130, 62/56 (Jess et al., 1996) A5968Rat HEK293 cDNA 80 (Mochizuki et al., 2005) CT (617–630)Rat Cos-7 cDNA 240, 120, 80, 60 NT (1–14)Rat CHO cDNA 90, 65, 60 (Ozaslan et al., 2003) CT (586–630)Rat Insect cDNA 60, 54 (Tate and Blakely, 1994) CT2/S365Rat BHK-21 cDNA 60, 55 (Tate et al., 2003) CT2Rat MEL cDNA 80, 60, 55Rat HEK293 cDNA 90Mouse HEK293 cDNA 80 (Hirano et al., 2005) CTMouse Brain Endogenous 70Mouse Macrophage Endogenous 70 (Rudd et al., 2005) AB1594Mouse Osteoclast Endogenous 62 (Bliziotes et al., 2006) AB1594Guinea pig Ileal mucosa Endogenous 75, 60 (Bian et al., 2007) CT (596–614)

Brain 70

Literature review indicating reported molecular weights for SERT endogenously expressed or generated from transfected cDNA in various tissues. ⁎Underlined data indicates majorspecies detected. †CT = C-terminal; NT = N-terminal; amino acid positions (where given) in brackets. ‡Suggested by authors to be non-specific. N.B. §Raised to a 15aa sequencebetween TMD 7 and 8. Studies reported where SERT protein band was indicated but with no molecular weight given are not included in this table.

76 A. Chamba et al. / Journal of Neuroimmunology 204 (2008) 75–84

Page 3: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

streptomycin. Confluent cultures of the colon carcinoma cell line Caco-2 were grown in high glucoseDMEM fromSigma-Aldrich (Dorset, U.K.)supplemented with 20% FCS, 1% non-essential amino acids (Sigma-Aldrich), glutamine, penicillin and streptomycin as above.

2.2. RNA isolation and RT-PCR

Total RNA was extracted from 107 cells using TRIzol reagent(Invitrogen) and precipitated with isopropanol, a further step toremove DNA was performed using the RNeasy mini kit (Qiagen, WestSussex, U.K.). Two micrograms of total RNA from each cell line wasreverse transcribed using Superscript II (Invitrogen). SERT primerswere generated to a conserved region of full-length human SERTmRNA (MWG Biotech, London, U.K.). SERT sense 5′-ATC ACG GCT GTGGTG GAT-3′ and SERT anti-sense 5′-ACT GAG TGA TGC CAT AGA ACCAA-3′ were used to amplify SERT transcripts. PCR conditions wereoptimised for SERTand the final programme usedwas 95 °C for 10minfollowed by 95 °C, 30 s; 55 °C, 30 s; 72 °C, 60 s for 38 cycles then 72 °C,10 min, using AmpliTaq Gold polymerase (Applera, Worrington, U.K.)and 1.5 mM Mg2+. The cDNA was also amplified for β-actin usingprimers, sense 5′-AGC GGG AAA TCG TGC GTG-3′ and anti-sense 5′-CAG GGT ACA TGG TGG TGC C-3′ for 25 cycles. PCR products wereseparated on 1.2% agarose gels containing ethidium bromide, DNA sizemarkers were run alongside samples. DNA from PCR gels wasextracted and purified using the QIAquick gel extraction kit (Qiagen),then sequenced by MWG Biotech.

2.3. Western blot and peptide blocking

SERT protein was analysed by Western immunoblotting. Briefly,107 cells were lysed in radioimmunoprecipitation assay (RIPA) buffercontaining 0.1% SDS/1% NP-40+protease inhibitors and a 50 μg totalprotein sample was subjected to 10% sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE). HEK-hSERT transfec-tants (10–25 μg) were used as a positive control. Proteins weretransferred to PVDF membranes and probed with 1:5000 dilution ofST51 (aa51–66) mouse monoclonal anti-human serotonin transporterantibody (Mab Technologies, Stone Mountain, GA) and 1:1000dilution of the C20 (aa611–630) goat polyclonal anti-human serotonintransporter antibody (Santa Cruz Biotechnology, CA). Blots werevisualised using an enhanced chemiluminescence method asdescribed previously (Serafeim et al., 2002). In order to confirmspecificity of the ST51 antibody, antibody was incubated with 5 foldexcess blocking peptide (PSPGAGDDTRHSIPAT) (Alta Bioscience,Birmingham, U.K.) in phosphate-buffered saline (PBS) for 2 h (mixedon a rotary shaker) at room temperature. The antibody–peptide mix-ture was made up to the appropriate antibody concentration andadded to membranes.

2.4. Measurement of intracellular SERT

For analysis of intracellular SERT 106 cells were fixed in 100 μl of 2%paraformaldehyde for 5 min on ice, washed in PBS then permeabilisedby resuspension in 500 μl of 0.1% saponin/PBS for 5 min on ice. Afterwashing in saponin/PBS, cells were incubatedwith either ST51 antibody(1:1000) or C20 (1:100) diluted in saponin/PBS for 30 min on ice. Cellswere washed in saponin/PBS then resuspended in appropriate second-ary antibody (goat anti-mouse PE or donkey anti-goat FITC) for 30 minon ice. Finally, cells were washed in PBS to reseal membranes thenresuspended in 5% NGS/1% formaldehyde/0.1% sodium azide/PBS andanalysed on a FACSCaliburflowcytometer (BectonDickinson, U.K.). IgG1

isotype controls at the same concentration as primary antibody wereused in control experiments. For peptide blocking, mouse SERT anti-body and peptide were mixed as described previously (Meredith et al.,2005b), then incubated with cells at the appropriate concentrationequivalent to that of antibody alone.

2.5. Biotinylation of cells

Biotinylation was performed using a modification of a methoddescribed elsewhere (Qian et al., 1997). Briefly, HEK-hSERT mono-layers or L3/Bcl2 (4×107) in suspension were gently washed with PBSthen treated with biotin-NHS; 10 mg/ml (Vector Laboratories,Peterborough, U.K.) in PBS containing 1 mM MgCl2 and 0.1 mMCaCl2, pH 7.4, at 4 °C for 1 h. Biotinylating reagents were removed bywashing cells with 100 mM glycine in PBS/Ca–Mg twice, the reactionwas quenched further by incubation with 100 mM glycine for 30 minon ice. Cells were then washed with cold PBS/Ca/Mg three timesbefore lysis with RIPA buffer containing protease inhibitors. Lysateswere centrifuged for 5 min at 13,000 rpm and supernatants wereincubated with monomeric avidin beads; 100 μl of beads/250 μl ofsupernatant (Pierce Biotechnology) for 2 h at room temperature orovernight at 4 °C. Beads werewashed three timeswith RIPA buffer andadsorbed proteins eluted with 50 μl (1×) gel loading buffer (6× buffer:62.5 mM Tris–HCL, pH 6.8, 20% glycerol, 2% SDS, 10% beta ME and5% bromophenol blue) for 30 min at room temperature on a rotaryshaker. Then a 10 μl sample from total cell lysate, lysate afterincubationwith avidin beads (non-biotinylated proteins) and the beadeluate (biotinylated proteins) was separated on 10% SDS-PAGE gels.To validate the surface localisation of biotinylated SERT, blots were re-probed with anti-β-actin antibody (Sigma; 1:10,000) followingVector-SG substrate blocking (Vector Laboratories) (Krajewski et al.,1996).

2.6. PNGase F and tunicamycin treatment

To remove N-linked glycans from SERT proteins, lysates wereprepared from HEK-hSERT and L3/Bcl-2 cells, then in the presenceof protease inhibitors enzymatically digested with PNGase F; 1 unit/100 μg protein (Roche Diagnostics) for 18 h at 37 °C. The reaction wasterminated by storage at −80 °C. To chemically block the productionof N-glycosylated proteins, cells were cultured in medium containingthe potent inhibitor tunicamycin which inhibits the reaction ofthe first step of N-linked oligosaccharide synthesis on glycoproteins,thus disrupting glycosylation of newly synthesised proteins; 10 μg/ml(Sigma) for 0, 24 and 48 h time points, after incubation cells wereharvested and lysates frozen. Frozen aliquots were thawed andsubjected to gel electrophoresis.

2.7. Immunocytochemistry and confocal staining

Typically, 106 cells were washed with cold PBS followed bycentrifugation. Cells were fixed for 10 minwith 2% paraformaldehyde,resuspended in cold PBS then spun at 1200 rpm for 5 min. 0.1%saponin/PBS was added to cells to permeabilise cell membranes, cellswere incubated for 5 min at RT then centrifuged for 5 min. ST51diluted in 0.1% saponin/PBS buffer (1:1000) was incubated with cellsfor 1 h at 4 °C, control experiments to determine antibody specificity/background stainingwere performed using isotype specific IgG1 at thesame concentration as primary antibody. Peptide blocking wasperformed using ST51/peptide mix as described above. The secondaryconjugate, goat anti-mouse TRITC or goat anti-mouse Alexa Fluor 488(1:50) (Invitrogen), was added after washing with PBS/saponin andincubated for 30 min at 4 °C. Following washing with saponin buffer,cells were incubated with 300 nM DAPI nuclear stain (Invitrogen) for5 min at room temperature. Co-staining for F-actin was performedwith phalloidin-TRITC at 250 ng/ml (Sigma) for 30 min following thesecondary antibody step. Cells were then washed with excess PBSto allow membranes to reseal, the final pellets were resuspended in asmall volume of PBS and an aliquot of cells spotted onto multiwellslides and left to air-dry. To prevent fading of the fluorochromes,DABCO (Sigma-Aldrich) was added to each cell spot, slides were keptin the dark at 4 °C. Staining was analysed using a laser-scanning

77A. Chamba et al. / Journal of Neuroimmunology 204 (2008) 75–84

Page 4: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

confocal microscopy system 510 (Zeiss, Hertfordshire, U.K.) usingArgon 488 and HeNe 543 sequential excitation.

3. Results

3.1. Expression of SERT in human cell lines of diverse peripheral origin

An amplified product for SERT was detected by PCR as expected inHEK293 cells transfected with hSERT (HEK-hSERT) but also in all othercell lines studied (including wild-type HEK293 cells) at the predictedsize of 223 bp (Fig. 1A). The nucleotide sequence of the PCR productwas sequenced from L3/Bcl-2 lymphocytes, CATCACCTGGCTTCTTTG-GATCCCTGGTCACCCTGACTTTTGGAGGGGCCTACGTGGTGAAGCTGCTG-GAGGAGTATGCCACGGGGCCCGCAGTGCTCACTGTCGCGCTGATCGAAG-CAGTC then compared with the BLAST database confirming that it isidentical (100% homology) to the human brain Na+/Cl−-dependenttransporter (GenBank=GenBank Accession Number X70697) (Leschet al., 1993). This transcript represents a sequence corresponding toaa474–510 contained within the region of SERT protein spanning TMD9–10. Though not quantitative, the most intense PCR product relativeto constant levels of β-actin was always generated from HEK-hSERTcells (Fig. 1A).

Having confirmed that all the cell lines selected for study ex-pressed SERT mRNA, SERT protein was analysed by Western blotanalysis. Given that some of the variability in reported SERTmolecularweight species (Table 1) could reflect the identification system used,we first compared six currently available anti-SERTantibodies for theirability to detect cell-extracted SERT resolved on reducing gels. Two ofthese failed, in our hands, to blot any detectable protein irrespective ofcell source fromwhich lysates were generated (including HEK-hSERT):Santa Cruz N-14 goat polyclonal antibody to SERT N-terminus;SERT11-A rabbit antibody to a 15aa sequence between TMD 7 and 8(Alpha Diagnostic). AB-N09 antibody (Advanced Targeting Systems),a mouse monoclonal generated against a peptide contained withinan extracellular domain of SERT (and recommended only for FACSstaining) universally blotted a discrete 96 kDa band in all cell lysates:wild-type HEK293 cells generating a band of equal intensity to thatfrom overexpressing HEK-hSERT transfectants. A similarly univer-sal blotting pattern was observed with Santa Cruz I-21 goat antibodyto C-terminusaa570–620, butwith the indiscriminate bandherehavingan apparent m.w. of 50 kDa (data with these antibodies not detailed).

The most reliable and consistent antibody in our hands was theST51 monoclonal antibody directed to aa51–66 near the human SERTN-terminus. Blottingwith ST51 demonstrated the expression of severalSERT bands in cell lysates: their precise representation and intensitydepending on cell of origin (Fig. 1B). In HEK-hSERT, a diffuse 85–95 kDabanddominated, consistentwith themobility ofN-glycosylatedSERT protein observed in previous studies using these cells (Rama-moorthy et al., 1998). HEK-hSERT also expressed SERT-reactive speciesat 70, 60 and 45 kDa. In the lymphoid cell lines, L3/Bcl-2, Daudi,RPMI8866, and Jurkat, only 70 and 60 kDa SERT bands were blottedwith ST51, the relative intensity of each of the bands varying with thecell line studied. In the non-lymphoid cell lines, Kelly, Caco-2, andparental HEK293, each of which similarly expressed the 70 and 60 kDaspecies, the additional faster migrating 45 kDa SERT band was alsodetected. To our knowledge this is the first time endogenous SERTprotein has been demonstrated in the wild-type HEK293 ‘epithelial’cell line (see Discussion) that is used widely in sert-transfectionstudies. Blocking experiments with a specific peptide immunogen(Fig. 1C) to the ST51 monoclonal antibody abolished all signal onWestern blots confirming the SERT immunoreactivity of each of thebands detected.

We also had reasonable success when blotting with ST C20, a goatpolyclonal antibody directed to SERT C-terminus aa611–630; however,the quality of blots was not ideal frequently showing weaker, morevariable bands and higher background levels than generated with

ST51 (results not detailed). Nevertheless we performed Westernblotting with this antibody on the same set of cells as above (with theexception of Caco-2 which became included for analysis onlyfollowing the report of SERT expression in these cells by Iceta et al.(28). Despite its limitations, C20 antibody was able to disclose SERTbands at 90 and 60 kDa in HEK-hSERT cells; in all other lines C20antibody detected only the 60 kDa band (as shown with the exampleof L3/Bcl-2 in blots performed on biotinylated fractions: see Fig. 4B);the 70 kDa species so clearly evident with ST51 antibody never beingobserved. Another report where C20 was used similarly found boththe diffuse 90 and discrete 60 kDa SERT species in HEK-hSERT cellswith platelets generating a dominant 100 kDa band (Frankhauseret al., 2006).

3.2. Quantitative assessment of SERT expression by FACS analysis

To provide a quantitative comparison of SERT expression in thedifferent lines, FACS-based analysis was used to measure total

Fig. 1. Expression of SERT in human cell lines. (A) RNA was extracted from each cellline using TRIzol then 2 μg RNAwere reverse transcribed to cDNA using Superscript II.SERT primers, sense 5′-ATC ACG GCT GTG GTG GAT-3′ and anti-sense 5′-ACT GAG TGATGC CATAGA ACC AA-3′were used to amplify SERT transcripts at 55 °C. A sample fromeach PCR reaction was visualised on a 1.2% agarose gel containing ethidium bromide,all cell lines revealed a single PCR product for SERT at 223 bp, a negative control sam-ple containing no RNAwas included in each PCR, as was a 100 bp DNA ladder. PCR forβ-actin is also shown to validate RNA integrity and equal loading. PCR images shownare representative of RT-PCR carried out on 3 separate RNA preparations. (B) SERTprotein expression was detected by Western blotting with anti-SERT ST-51 (1:5000)mouse monoclonal antibody in cell lysates (50 μg), a stable line of HEK293 cellscarrying full-length human SERT (HEK-hSERT) was used as a positive control on blots(10–25 μg). A and B are representative of three independent experiments. (C) Peptideblocking shows detection of SERT in HEK-hSERT and L3/Bcl-2 lysates with ST51±specific blocking peptide (PSPGAGDDTRHSIPAT).

78 A. Chamba et al. / Journal of Neuroimmunology 204 (2008) 75–84

Page 5: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

immunoreactive SERT in permeabilised cells (Fig. 2). As anticipated,HEK-hSERT cells showed high level staining with ST51 monoclonalantibody that was reversed in the presence of inhibitory peptide(Fig. 2A). The levels of ST51-reactive SERTcarried endogenously in othercell lines, though readily detectable by FACS (and again reversed bypeptide immunogen), were considerably lower than inHEK-hSERTcells.Staining was also performed with the C20 polyclonal antibody (Fig. 2B),again showing high mean fluorescence intensity (MFI) values in HEK-hSERT but significantly lower (although clearly detectable) levels in theother cells. The level of immunoreactive SERT contained endogenouslywithin wild-type HEK293 epithelial cells represented 10% and 14% ofthat generated in transfected HEK-hSERT as measured with ST51 andC20 antibody, respectively. The lowest levels of SERT, as measured witheither antibody, were seen in the Daudi B-lymphoma line and theRPMI8866 B-lymphoblastoid cell line (Fig. 2B); L3/Bcl-2 B-lymphomacells, Jurkat T cells and Kelly neuroblastoma/sympathoadrenal cellscarried levels of SERT similar to wild-type HEK239.

3.3. Contribution of N-glycosylation to the different SERT protein speciesdetected

The multiple protein bands detected with ST51 prompted us toexamine the presence of SERT N-linked glycosylation (Fig. 3).Enzymatic removal of N-linked glycans by PNGase F produced altered

mobility of the diffuse 85–95 kDa band in extracts of HEK-hSERT, to abroad band appearing to contain the 70 and 60 kDa bands whenblotted with ST-51 (Fig. 3A): the resolution of these two speciesbecoming more evident on shorter exposure times of the Westernblots (Fig. 3A; lower panel). Our results are in agreement withprevious studies indicating that the 85–95 kDa band representsprecursor protein which has been heavily glycosylated by post-translational modification (Ramamoorthy et al., 1998). Importantly,the observation that the broad 85–95 kDa band produces distinct 70and 60 kDa species on deglycosylation would imply that these lowermolecular weight products, which are also represented but at a lowerlevel in pre-treatment samples, are not simple degradation polypep-tides but rather represent stable non-glycosylated forms of thetransporter. Similarly, the generation of the two non-glycosylatedbands from transfected hSERT strongly supports the SERT nature ofthe same 70 and 60 kDa products found endogenously in the differentcell lines of lymphoid and non-lymphoid origin. In L3/Bcl-2 lymphomacells which expressed only the 70 and 60 kDa SERT protein bandstherewas nomeasurable shift in themolecular sizes following PNGaseF treatment (Fig. 3A), implying again that these SERT species havenot been modified by glycosylation. Similarly, wild-type HEK293extracts shown on blots for comparison, also produced no alterationin bands following deglycosylation treatment suggesting these non-glycosylated SERT forms are not restricted to lymphoid cells. The

Fig. 2. SERT FACS analysis. (A) Cells were stained for total cellular SERT with ST-51, briefly cells were permeabilised, incubated with ST51 (1/1000) for 30 min followed by goat anti-mouse PE. Samples were analysed on a FACS Calibur flow cytometer. Panel shows the staining profiles of cell lines, histograms are shown as specific ST51 stain (open) and inhibitionwith specific peptide (bold). Negative controls were performed with species specific IgG (shaded). (B) Cells were stained with ST51 as above, or with C20 (1/100) for 30 min followedby donkey anti-goat FITC. Mean fluorescence intensity (MFI, arbitrary units) from three independent experiments±SE is presented.

79A. Chamba et al. / Journal of Neuroimmunology 204 (2008) 75–84

Page 6: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

discrete 45 kDa band present in HEK-hSERT (and in the non-lymphoidcell lines endogenously) also showed no shift in its size or intensityfollowing deglycosylation of the sample.

Confirmation of the glycosylation state of SERT was obtained byculturing cells in the presence of tunicamycin (Fig. 3B). In HEK-hSERT,adding tunicamycin prevented formation of N-glycans andmaturationof the large 85–95 kDa band within 24 h, these findings confirmingthose from the PNGase F experiments above. Also in agreement withthe PNGase F experiments, tunicamycin treatment did not inhibit thegeneration of the 70 and 60 kDa bands in either HEK-hSERT or L3/Bcl-2 lymphoma cells.

3.4. Localisation of SERT endogenous to lymphoid cells

Biotinylation experiments demonstrated SERT immunoreactivityin both plasma membrane and intracellular (non-biotinylated)fractions of HEK-hSERT and L3/Bcl-2 (Fig. 4). In HEK-hSERT cells themajority of SERT isolated from themembrane surfacewas representedby the 85–95 kDa glycosylated form (Fig. 4A); an approximately equalproportion of the SERT was intracellular in these cells. The 45 kDaHEK293 SERT species was not accessible for biotinylation. In L3/Bcl-2lymphoid cells, the non-glycosylated 60 kDa species was themajor – ifnot only – form detected in membrane extracts (Fig. 4A). Both 60 and70 kDa forms were expressed in the intracellular fractions of thesecells. Blots were re-probed for non-surface-exposed β-actin tovalidate the rigour of the surface biotinylation procedure (Fig. 4A;lower panel). Samples of HEK-hSERT and L3/Bcl-2 total, surface andintracellular extracts used in Fig. 4A were then blotted with ST C20

(Fig. 4B); again the C20 antibody predominantly blotted a broad90 kDa surface and intracellular form in hSERT-transfected HEK293cells but in the lymphoid cells a single band of 60 kDa was blotted inall three fractions.

To confirm the glycosylation state of membrane expressed SERT,biotinylated surface extracts were treated with PNGase F as for Fig. 3.In HEK-hSERT cells, surface available 85–95 kDa SERT again shiftedto a heavy broad band of substantially lower molecular weight onremoving N-glycans. For L3/Bcl-2 cells, there was also again nodiscernible change in the mobility of the 60 kDa surface protein onPNGase F treatment (Fig. 4C). The findings reveal that a 60 kDa SERTprotein traffics to the surfacemembrane of lymphoid cells despite lackof modification by N-glycans.

Fig. 3. Deglycosylation of SERT. (A) Total protein extracts from L3/Bcl-2 (50 μg) and HEK-hSERT (25 μg) were incubated with PNGase F (1 unit) for 18 h at 37 °C to remove N-linkedglycans, then subjected to Western blotting and probed with ST51 SERT antibody, PNGaseF treated lysates (+) were compared with corresponding untreated lysates (−). Two expo-sures of the same blot are shown at 20 and 30 s. (B) Cells were cultured in the presenceof tunicamycin (10 μg/ml at 37 °C), a potent chemical inhibitor ofN-glycosylation, for 0, 24and 48 h time points. After incubation cells were harvested and lysates prepared forblotting, sample amountswere loadedas above.Western blots shownare representative ofat least three separate experiments.

Fig. 4.Distribution of SERT in lymphocytes. (A and B) L3/Bcl-2 and HEK-hSERTcells weretreated with NHS-biotin to label surface SERT as described in Materials and methodsthen intracellular non-biotinylated (non-b) and surface biotinylated (surface) proteinswere purified from each cell. A small aliquot of total protein extract was retained fromeach experiment to load on gels. Awash sample (wash) from each extractionwas loadedon gels to ensure removal of all non-biotinylated proteins from beads before extractionof surface protein. Purified proteins were probed with either anti-SERT ST51 (A) or anti-SERT C20 (B), subsequently blots were re-probed with anti-β-actin (1:10,000)monoclonal antibody after SG substrate blocking to validate surface biotinylation aswell as equal protein loading. Immunoblots show total (50 μg), non-biotinylated (50 μg)and surface (10 μl sample) proteins and are representative of at least 3 independentexperiments. (C) PNGase F treatment of surface SERT. Surface biotinylated protein wasextracted from L3/Bcl-2 and HEK-hSERT then treated with PNGase F and blotted withST51, samples of total protein from each cell line are shown for comparison.

80 A. Chamba et al. / Journal of Neuroimmunology 204 (2008) 75–84

Page 7: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

To further investigate cellular localisation of SERT in HEK-hSERTand L3/Bcl-2 cells we performed immunofluorescence stainingfollowed by confocal laser-scanning microscopy (Fig. 5). To establishthe validity of this approach, staining was first performed with theappropriate (IgG1) isotype control, the ST51 SERT antibody, and theantibody blocked with peptide. No background staining was observedand SERT staining was extinguished by blocking peptide in both HEK-hSERT and L3/Bcl-2 cells; confirming the specificity of the reagentsused (Fig. 5A).

HEK-hSERT cells stained strongly for SERT and a significantproportion of this staining co-localised with F-actin at the cell surface(Fig. 5B) as indicated by the green (SERT) and red (F-actin) signalsmerging to generate a yellow signal; this is in full agreement with theabove biotinylation studies indicating that a substantial fraction of theheavily glycosylated SERT in these cells is expressed at the cell surface.By contrast and in agreement with both FACS and blotting data,

staining for the endogenous SERT protein contained in L3/Bcl-2 cellswas appreciably weaker. These lower concentrations of SERTappearedto be largely localised to intracellular compartments, no strong co-localisation with F-actin being evident from these experiments. Thisresult is consistent with our finding that only a small fraction of thetotal endogenous SERT present in L3/Bcl-2 cells could be accessed atthe outer surface membrane through biotinylation.

4. Discussion

SERT is now recognised as being expressed in cell types of diverseperipheral origin including platelets, lymphocytes and placental cells;thesefindings indicating a role for thehigh affinity serotonin transporterin regulating cellular function outside of the central nervous system.Despite a largenumberof studies characterisingSERTat theprotein level(Table 1) no consensus exists regarding its size and isoforms within or

Fig. 5. SERT immunostaining. (A) L3/Bcl-2 and HEK-hSERT were immunostained for SERT as described in Materials and methods, peptide blocking experiments were performed oneach cell line. SERT staining with ST51 (1:1000) was visualised with goat anti-mouse TRITC (SERT; red signal), staining of cell nuclei with DAPI is shown as blue signal, controls wereperformed with mouse IgG1. Co-localisation (B) shows three different fields for each cell line, staining was performed using ST51 antibody and visualised with goat anti-mouse 488conjugate (SERT; green signal), F-actinwas stained with phalloidin-TRITC (Sigma-Aldrich) (F-actin; red signal) to define the cell membrane, merged view shows the co-localisation ofSERT and F-actin as yellow fluorescence. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

81A. Chamba et al. / Journal of Neuroimmunology 204 (2008) 75–84

Page 8: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

outside the CNS, the extent to which post-translational modificationdetermines these, or the contribution of N-glycosylation to its surfaceexpression— particularly in endogenous backgrounds. Our own interestin this field came from identifying a functional serotonin transporterin human Burkitt's lymphoma cells (Serafeim et al., 2002) then morewidely in a range of B-cell malignancies as well as in normal B-cellsupon their activation (Meredith et al., 2005b; Serafeim et al., 2003).As SERT offers a potential therapeutic target in this context we felt itimportant to clarify the exact nature of the protein at the lymphoid cellsurface, comparing it to that found on other peripheral cell types and –as reference – to the well studied product generated in HEK293 cellstransfectedwith full-length human sert cDNA cloned from the placentaltrophoblastic cell line JAR (Ramamoorthy et al., 1993).

All the cell lines incorporated into this study on the basis ofreported SERT protein expression contained readily detectable SERTtranscripts, the sequence of which was confirmed as being identical tothat contained within the cDNA from JAR cells. Completely unex-pected at the time was the finding that wild-type HEK293 cells –included simply as controls to the HEK-hSERT transfectants – alsoexpressed SERT transcripts constitutively. This observation carriedthrough to the protein level. It has subsequently been brought to ourattention that using the Clontech gene array platform, Shaw et al.(2002) identified 5HTT (SERT) as the single most abundant mRNAdetected in (wild-type) HEK293 cells: together with a surprisinglylarge representation of other transcripts (and proteins) usuallyassociated with the CNS, this observation led the authors to concludethat a rare neuronal cells within the embryonic kidney was targetedfor transformation by adenovirus to generate the stable line classicallyconsidered to be of epithelial origin. The three major SERT proteinspecies carried in parental HEK293 cells similarly appeared in theCaco-2 endothelial line and Kelly sympathoadrenal cells. Of these, the60 and 70 kDa species were shared by all four lymphoid cell linesstudied: the remaining 45 kDa band being essentially restricted to thenon-lymphoid cells. A similar immunoreactive protein of 43 kDa hasbeen shown previously in human platelet extracts (Belous et al., 2001)and more recently a 40 kDa SERT band was detected in rat intestinalmucosa and rat brain (Iceta et al., 2006) suggesting that this SERTband may not be the result of site-specific endoproteolytic cleavage asconcluded by other workers (Dmitriev et al., 2004).

As expected the 45, 60 and 70 kDa SERT species carriedendogenously in wild-type HEK293 cells also appeared in HEK-hSERTtransfectants; and at roughly comparable level. Relatively, however,these registered as minor components only with the previouslyreported broad 85–95 kDa diffuse band dominating blots generatedfrom the sert-transfectants. Importantly, when using the ST51 mono-clonal antibody for detection, all SERT-reactive bands – as representedinHEK-hSERT – disappeared on blocking immunoreactivitywith excessspecific peptide. Despite the range of sizes reported for SERT protein ashighlighted in Table 1, there is a trend for the major species in HEK-hSERT to be of higher m.w. than that found endogenously; or indeedwhere sert cDNA has been introduced into other cellular backgrounds,such as HeLa cells. A clear exception is indicated by studies on plateletswhere an approximate 100 kDa SERT form appears to dominate. Ourown experience regarding platelet SERT generated inconsistent dataeven when blotting with ST51. In two of three platelet preparations alarge diffuse band similar in size to that of the 85–95 kDa speciesdetected in HEK-hSERT was the most abundant found with the 60 and/or 70 kDa species appearing irregularly, all of these again specificallypeptide-blocked and with the 45 kDa species never detected (datanot detailed).

Following previous studies we addressed the contribution ofglycosylation to the heterogeneity in SERT products observed here.SERT is recognised as displaying cell-type dependent glycosylationpatterns: for example endogenous SERT in human brain and plateletsreported at 76 and 94 kDa respectively (Qian et al., 1995) whendeglycosylated generated a single species of 56 kDa. Similarly, in CHO

cells transfected with rat SERT, glycosylated forms of 90, 65 and60 kDa were reduced to single band of non-glycosylated protein at50 kDa (Ozaslan et al., 2003). Another study of rat brain SERTsuggested a 70 kDa glycosylated form is expressed which is convertedto 50 kDa on deglycosylation (Wang et al., 2005). First we confirmedthat the broad 85–95 kDa SERT band generated from HEK-hSERT cellsis heavily N-glycosylated: both PNGase F and tunicamycin treatmentresulting in its disappearance. Here for the first time we have shownthe resolution of the diffuse high m.w. species to two discrete bandson removing N-glycans the mobilities of which correspond preciselyto those of the 60 and 70 kDa species found constitutively in non-transfected cells: lymphoid and non-lymphoid. Correspondingly, themobility of these endogenous species – including the 45 kDa productin non-lymphoid cells – remained unchanged on deglycosylation. Wetherefore conclude that the 60 and 70 kDa SERT species carriedendogenously by the range of peripheral cells studied here are non-glycosylated proteins that in discrete scenarios – for example, whenoverexpressed in HEK293 or, constitutively, in platelets – are subjectto N-glycosylation. Clearly it is not the cellular background per se thatimposes this difference. One possibility is that high level expression asseen with both HEK-hSERT and platelets may of itself encourageglycosylation of the transporter.

We next asked whether the N-glycan-deficient SERT associatedwith peripheral cells was actually capable of reaching the surface andthereby access exogenous ligand(s). The result appeared unequivocal:the 60 kDa, but not 70 kDa, SERT species was available at the surfaceto biotin as modelled in L3/Bcl-2 lymphoma cells. The proportion ofSERTachieving this was small however, certainly by comparison to theefficient representation of glycosylated SERT at the surface of HEK-hSERT cells and as indicated by the primarily intracytoplasmicdistribution of SERT immunoreactivity in the lymphoid cells. Reportson related transporters have highlighted the importance of glycosyla-tionboth to trafficking and function (Keynan et al.,1992;Melikian et al.,1996). However, another study concludes that while N-glycansimprove the ability of the SERT polypeptide to fold into a functionaltransporter they are not required for the transmembrane transport ofserotonin (Tate et al., 2003). On a background of Sf9 insect cells, Tateand Blakely (1994) concluded that while non-glycosylated SERT was20-fold less efficient at trafficking to the surface, the small proportionof core protein that did arrive showed unchanged ligand-bindingactivity and similarly unaltered 5-HT transport capacity. These reportsand the data presented here when coupled with our earlier finding ofB-lymphoma cells actively engaging in 5-HT transport (Serafeim et al.,2002) indicate that the 60 kDa non-glycosylated membrane SERTis indeed a functional transporter: a finding supported by evidencethat wild-type HEK293 cells (which we show fail to glycosylate theirendogenous SERT) is fully competent for 5-HT uptake (Shaw et al.,2002). Similar conclusions have been reached for the related SLC6 genefamily member DAT where removal of the canonical N-glycosylationsites fails to abolish membrane trafficking or dopamine uptake butdoes result in a reduced representation of the transporter at the cellsurface (Li et al., 2004).

Finally, it is of interest to speculate on the nature of the 60 kDaversus 70 kDa SERT carried endogenously in all SERT-expressing cellsso far studied — irrespective of their lineage. Different isoforms couldbe attributed to alternative spliced transcripts as exemplified by theCRF1 receptor (Pisarchik and Slominski, 2004). However, in the presentstudy apparent SERT isoforms will not arise from differential splicingas both are generated from sert cDNA as indicated by their increasein abundance following deglycosylation of the over expressed productfrom HEK-hSERT cells. Qian et al. (1995) on noting a smaller but con-sistent difference (2 kDa) in mobility between N-deglycosylatedrat SERT derived from endogenous sources or transfected cellsposited O-glycosylation, sulfation, acylation and/or phosphorylationas candidate modifications to contribute additional heterogeneity. Theinvolvement of O-glycosylation and other modifications in human

82 A. Chamba et al. / Journal of Neuroimmunology 204 (2008) 75–84

Page 9: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

lymphocytic SERT could be explored further by enzymatic removal oftarget functional groups and proteomic analysis. A clue to the pre-sumed modification might come from the inability of C20 antibody toreact with 70 kDa human SERT in our studywhile readily detecting the60 kDa form:modificationwithin the C-terminus stretch of aa611–630to which the antibody is raised potentially extinguishing epitoperecognition or altering the antibody binding ability. The presence oftwo threonines and a serinewithin this short sequence prompted us toexplore the known PKC-dependent phosphorylation of SERT (Rama-moorthy et al., 1998) as a likely explanation. However, a number ofapproaches aimed to address this (e.g. phorbol ester-driven phos-phorylation, enzymatic dephosphorylation) were without successleaving the issue currently unresolved.

The clearer understanding of the nature of SERT expressed andfunctioning endogenously on (non-platelet) cells in the peripheryshould aid future study of its precise role in the tissues where it isincreasingly being found and, in select scenarios, – as with the case ofB-cell malignancy – assist rational drug design where the transporteroffers itself as a potential target for therapeutic attack. Furthermore,compared to brain, peripheral cells – as exemplified by lymphocytes –provide a tractable source of both SERT protein and transcripts whichcould be exploited as surrogates for assessing andmonitoring functionand disturbances associated with the CNS (Meredith et al., 2005a).

Acknowledgements

The work described in this paper was supported by a project grantfrom the Leukemia Research Fund (U.K.). We wish to thank RandyBlakely (Vanderbilt University, Nashville, TN) for generously providingHEK-hSERT cells and advice in the early part of this study.

References

Balkovetz, D.F., Tiruppathi, C., Leibach, F.H., Mahesh, V.B., Ganapathy, V., 1989. Evidencefor an imipramine-sensitive serotonin transporter in human placental brush-border membranes. J. Biol. Chem. 264, 2195–2198.

Barkan, T., Gurwitz, D., Levy, G., Weizman, A., Rehavi, M., 2004. Biochemical andpharmacological characterization of the serotonin transporter in human peripheralblood lymphocytes. Eur. Neuropsychopharmacol. 14, 237–243.

Belous, A.R., Ramamoorthy, S., Blakely, R.D., Factor, M.I., Dupin, A.M., Katasonov, A.B.,Lozier, R.H., Beniashvili, A.G., Morozova, M.A., Brusov, O.S., 2001. The state of theserotonin transporter protein in the platelets of patients with somatoform[correction of somatiform] disorders. Neurosci. Behav. Physiol. 31, 185–189.

Bian, X., Patel, B., Dai, X., Galligan, J.J., Swain, G., 2007. High mucosal serotoninavailability in neonatal guinea pig ileum is associated with low serotonintransporter expression. Gastroenterology 132, 2438–2447.

Bliziotes, M., Eshleman, A., Burt-Pichat, B., Zhang, X.W., Hashimoto, J., Wiren, K., Chenu,C., 2006. Serotonin transporter and receptor expression in osteocytic MLO-Y4 cells.Bone 39, 1313–1321.

Cai, G., Salonikidis, P.S., Fei, J., Schwarz, W., Schulein, R., Reutter, W., Fan, H., 2005. Therole of N-glycosylation in the stability, trafficking and GABA-uptake of GABA-transporter 1. Terminal N-glycans facilitate efficient GABA-uptake activity of theGABA transporter. FEBS J. 272, 1625–1638.

Dmitriev, A.D., Pavlova, E.V., Factor, M.I., Segal, O.L., Massino, Y.S., Dobrohotov, I.V.,Smirnova, M.B., Hwun, D.E., Yakovleva, D.A., Kolyaskina, G.I., Brusov, O.S., 2004.Analysis of serotonin transporter in human platelets by immunoblotting using site-specific antibodies. Biochemistry (Mosc.) 69, 629–641.

Faraj, B.A., Olkowski, Z.L., Jackson, R.T., 1994. Expression of a high-affinity serotonintransporter in human lymphocytes. Int. J. Immunopharmacol. 16, 561–567.

Frankhauser, P., Grimmer, Y., Bugert, P., Deuschle, M., Schmidt, M., Schloss, P., 2006.Characterization of the neuronal dopamine transporter DAT in human bloodplatelets. Neurosci. Lett. 399, 197–201.

Gordon, J., Barnes, N.M., 2003. Lymphocytes transport serotonin and dopamine: agonyor ecstasy? Trends Immunol. 24, 438–443.

Hirano, K., Seki, T., Sakai, N., Kato, Y., Hashimoto, H., Uchida, S., Yamada, S., 2005. Effectsof continuous administration of paroxetine on ligand binding site and expression ofserotonin transporter protein in mouse brain. Brain Res. 1053, 154–161.

Hranilovic, D., Herak-Kramberger, C.M., Cicin-Sain, L., Sabolic, I., Jernej, B., 2001.Serotonin transporter in rat platelets. Level of protein expression underliesinherited differences in uptake kinetics. Life Sci. 69, 59–65.

Iceta, R., Mesonero, J.E., Aramayona, J.J., Alcalde, A.I., 2006. Molecular characterizationand intracellular regulation of the human serotonin transporter in Caco-2 cells.J. Physiol. Pharmacol. 57, 119–130.

Jayanthi, L.D., Samuvel, D.J., Blakely, R.D., Ramamoorthy, S., 2005. Evidence for biphasiceffects of protein kinase C on serotonin transporter function, endocytosis, andphosphorylation. Mol. Pharmacol. 67, 2077–2087.

Jess, U., Betz, H., Schloss, P., 1996. The membrane-bound rat serotonin transporter,SERT1, is an oligomeric protein. FEBS Lett. 394, 44–46.

Keynan, S., Suh, Y.J., Kanner, B.I., Rudnick, G., 1992. Expression of a cloned gamma-aminobutyric acid transporter in mammalian cells. Biochemistry 31, 1974–1979.

Krajewski, S., Zapata, J.M., Reed, J.C., 1996. Detection of multiple antigens on westernblots. Anal. Biochem. 236, 221–228.

Launay, J.M., Geoffroy, C., Mutel, V., Buckle, M., Cesura, A., Alouf, J.E., Da Prada, M., 1992.One-step purification of the serotonin transporter located at the human plateletplasma membrane. J. Biol. Chem. 267, 11344–11351.

Lesch, K.P., Wolozin, B.L., Estler, H.C., Murphy, D.L., Riederer, P., 1993. Isolation of a cDNAencoding the human brain serotonin transporter. J. Neural. Transm. Gen. Sect. 91,67–72.

Li, L.B., Chen, N., Ramamoorthy, S., Chi, L., Cui, X.N., Wang, L.C., Reith, M.E., 2004. The roleof N-glycosylation in function and surface trafficking of the human dopaminetransporter. J. Biol. Chem. 279, 21012–21020.

Little, K.Y., Zhang, L., Cook, E., 2006. Fluoxetine-induced alterations in human plateletserotonin transporter expression: serotonin transporter polymorphism effects.J. Psychiatry Neurosci. 31, 333–339.

Malison, R.T., Price, L.H., Berman, R., van Dyck, C.H., Pelton, G.H., Carpenter, L., Sanacora,G., Owens, M.J., Nemeroff, C.B., Rajeevan, N., Baldwin, R.M., Seibyl, J.P., Innis, R.B.,Charney, D.S., 1998. Reduced brain serotonin transporter availability in majordepression as measured by [123I]-2 beta-carbomethoxy-3 beta-(4-iodophenyl)tropane and single photon emission computed tomography. Biol Psychiatry 44,1090–1098.

Marcusson, J.O., Ross, S.B., 1990. Binding of some antidepressants to the 5-hydroxytryptamine transporter in brain and platelets. Psychopharmacology (Berl)102, 145–155.

Melikian, H.E., Ramamoorthy, S., Tate, C.G., Blakely, R.D., 1996. Inability to N-glycosylatethe human norepinephrine transporter reduces protein stability, surface trafficking,and transport activity but not ligand recognition. Mol. Pharmacol. 50, 266–276.

Meredith, E.J., Chamba, A., Holder, M.J., Barnes, N.M., Gordon, J., 2005a. Close encounters ofthe monoamine kind: immune cells betray their nervous disposition. Immunology115, 289–295.

Meredith, E.J., Holder, M.J., Chamba, A., Challa, A., Drake-Lee, A., Bunce, C.M., Drayson,M.T., Pilkington, G., Blakely, R.D., Dyer, M.J., Barnes, N.M., Gordon, J., 2005b.The serotonin transporter (SLC6A4) is present in B-cell clones of diverse malig-nant origin: probing a potential anti-tumor target for psychotropics. FASEB J. 19,1187–1189.

Mochizuki, H., Amano, T., Seki, T., Matsubayashi, H., Mitsuhata, C., Morita, K., Kitayama,S., Dohi, T., Mishima, H.K., Sakai, N., 2005. Role of C-terminal region in the functionalregulation of rat serotonin transporter (SERT). Neurochem. Int. 46, 93–105.

Najib, A., Pelliccioni, P., Gil, C., Aguilera, J., 2000. Serotonin transporter phosphorylationmodulated by tetanus toxin. FEBS Lett. 486, 136–142.

Ni, W., Thompson, J.M., Northcott, C.A., Lookingland, K., Watts, S.W., 2004. The serotonintransporter is present and functional in peripheral arterial smooth muscle.J. Cardiovasc. Pharmacol. 43, 770–781.

Ozaslan, D., Wang, S., Ahmed, B.A., Kocabas, A.M., McCastlain, J.C., Bene, A., Kilic, F.,2003. Glycosyl modification facilitates homo- and hetero-oligomerization of theserotonin transporter. A specific role for sialic acid residues. J. Biol. Chem. 278,43991–44000.

Pisarchik, A., Slominski, A., 2004. Molecular and functional characterization of novelCRFR1 isoforms from the skin. Eur. J. Biochem. 271, 2821–2830.

Pizzinat, N., Girolami, J.P., Parini, A., Pecher, C., Ordener, C., 1999. Serotonin metabolismin rat mesangial cells: involvement of a serotonin transporter and monoamineoxidase A. Kidney Int. 56, 1391–1399.

Purselle, D.C., Nemeroff, C.B., 2003. Serotonin transporter: a potential substrate in thebiology of suicide. Neuropsychopharmacology 28, 613–619.

Qian, Y., Melikian, H.E., Rye, D.B., Levey, A.I., Blakely, R.D., 1995. Identification andcharacterization of antidepressant-sensitive serotonin transporter proteins usingsite-specific antibodies. J. Neurosci. 15, 1261–1274.

Qian, Y., Galli, A., Ramamoorthy, S., Risso, S., DeFelice, L.J., Blakely, R.D., 1997. Proteinkinase C activation regulates human serotonin transporters in HEK-293 cells viaaltered cell surface expression. J. Neurosci. 17, 45–57.

Ramamoorthy, S., Blakely, R.D., 1999. Phosphorylation and sequestration of serotonintransporters differentially modulated by psychostimulants. Science 285, 763–766.

Ramamoorthy, S., Bauman, A.L., Moore, K.R., Han, H., Yang-Feng, T., Chang, A.S.,Ganapathy, V., Blakely, R.D., 1993. Antidepressant- and cocaine-sensitive humanserotonin transporter: molecular cloning, expression, and chromosomal localiza-tion. Proc. Natl. Acad. Sci. U. S. A. 90, 2542–2546.

Ramamoorthy, S., Giovanetti, E., Qian, Y., Blakely, R.D., 1998. Phosphorylation andregulation of antidepressant-sensitive serotonin transporters. J. Biol. Chem. 273,2458–2466.

Rothman, R.B., Baumann, M.H., 2003. Monoamine transporters and psychostimulantdrugs. Eur. J. Pharmacol. 479, 23–40.

Rothman, R.B., Jayanthi, S., Wang, X., Dersch, C.M., Cadet, J.L., Prisinzano, T., Rice, K.C.,Baumann, M.H., 2003. High-dose fenfluramine administration decreases serotonintransporter binding, but not serotonin transporter protein levels, in rat forebrain.Synapse 50, 233–239.

Rotondo, A., Giannaccini, G., Betti, L., Chiellini, G., Marazziti, D., Martin, C., Lucacchini, A.,Cassano, G.B., 1996. The serotonin transporter from human brain: purification andpartial characterization. Neurochem. Int. 28, 299–307.

Rudd, M.L., Nicolas, A.N., Brown, B.L., Fischer-Stenger, K., Stewart, J.K., 2005. Peritonealmacrophages express the serotonin transporter. J. Neuroimmunol. 159, 113–118.

Rudnick, G., Wall, S.C., 1992. The molecular mechanism of “ecstasy” [3,4-methylene-dioxy-methamphetamine (MDMA)]: serotonin transporters are targets for MDMA-induced serotonin release. Proc. Natl. Acad. Sci. U. S. A. 89, 1817–1821.

83A. Chamba et al. / Journal of Neuroimmunology 204 (2008) 75–84

Page 10: Characterisation of the endogenous human peripheral ...celentyx.com/uploads/3/5/1/2/3512419/chamba_et_al_2008.pdfCharacterisation of the endogenous human peripheral serotonin transporter

Serafeim, A., Grafton, G., Chamba, A., Gregory, C.D., Blakely, R.D., Bowery, N.G., Barnes,N.M., Gordon, J., 2002. 5-Hydroxytryptamine drives apoptosis in biopsylike Burkittlymphoma cells: reversal by selective serotonin reuptake inhibitors. Blood 99,2545–2553.

Serafeim, A., Holder, M.J., Grafton, G., Chamba, A., Drayson,M.T., Luong, Q.T., Bunce, C.M.,Gregory, C.D., Barnes, N.M., Gordon, J., 2003. Selective serotonin reuptake inhibitorsdirectly signal for apoptosis in biopsy-like Burkitt lymphoma cells. Blood 101,3212–3219.

Shaw, G., Morse, S., Ararat, M., Graham, F.L., 2002. Preferential transformation of humanneuronal cells by human adenoviruses and the origin of HEK 293 cells. FASEB J. 16,869–871.

Shively, C.A., Mirkes, S.J., Lu, N.Z., Henderson, J.A., Bethea, C.L., 2003. Soy and socialstress affect serotonin neurotransmission in primates. Pharmacogenomics J. 3,114–121.

Tate, C.G., Blakely, R.D.,1994. The effect of N-linked glycosylation on activity of theNa(+)-and Cl(−)-dependent serotonin transporter expressed using recombinant baculo-virus in insect cells. J. Biol. Chem. 269, 26303–26310.

Tate, C.G., Haase, J., Baker, C., Boorsma, M., Magnani, F., Vallis, Y., Williams, D.C., 2003.Comparison of seven different heterologous protein expression systems for theproduction of the serotonin transporter. Biochim. Biophys. Acta 1610, 141–153.

Torres, G.E., Gainetdinov, R.R., Caron, M.G., 2003. Plasma membrane monoaminetransporters: structure, regulation and function. Nat. Rev., Neurosci. 4, 13–25.

Tsao, C.W., Lin, Y.S., Cheng, J.T., Chang,W.W., Chen, C.L., Wu, S.R., Fan, C.W., Lo, H.Y., 2006.Serotonin transporter mRNA expression is decreased by lamivudine and ribavirinand increased by interferon in immune cells. Scand. J. Immunol. 63, 106–115.

Wade, P.R., Chen, J., Jaffe, B., Kassem, I.S., Blakely, R.D., Gershon, M.D., 1996. Localizationand function of a 5-HT transporter in crypt epithelia of the gastrointestinal tract.J. Neurosci. 16, 2352–2364.

Wang, X., Baumann, M.H., Xu, H., Morales, M., Rothman, R.B., 2005. (+/−)-3,4-Methylenedioxymethamphetamine administration to rats does not decrease levelsof the serotonin transporter protein or alter its distribution between endosomesand the plasma membrane. J. Pharmacol. Exp. Ther. 314, 1002–1012.

Wersinger, C., Rusnak, M., Sidhu, A., 2006. Modulation of the trafficking of the humanserotonin transporter by human alpha-synuclein. Eur. J. Neurosci. 24, 55–64.

Xie, T., Tong, L., McLane, M.W., Hatzidimitriou, G., Yuan, J., McCann, U., Ricaurte, G.,2006. Loss of serotonin transporter protein after MDMA and other ring-substitutedamphetamines. Neuropsychopharmacology 31, 2639–2651.

Zhou, F.C., Xu, Y., Bledsoe, S., Lin, R., Kelley, M.R., 1996. Serotonin transporter antibodies:production, characterization, and localization in the brain. Brain Res. Mol. Brain Res.43, 267–278.

84 A. Chamba et al. / Journal of Neuroimmunology 204 (2008) 75–84