twik-relatedacid-sensitivek channel1(task1)andtask3 ... · for evaluation of task1/3 antibody...

13
TWIK-related Acid-sensitive K Channel 1 (TASK1) and TASK3 Critically Influence T Lymphocyte Effector Functions * Received for publication, January 24, 2008, and in revised form, March 6, 2008 Published, JBC Papers in Press, March 28, 2008, DOI 10.1074/jbc.M800637200 Sven G. Meuth ‡1,2 , Stefan Bittner ‡1,3 , Patrick Meuth § , Ole J. Simon , Thomas Budde , and Heinz Wiendl From the Department of Neurology, University of Wuerzburg, Josef-Schneider-Str. 11, 97080 Wuerzburg, the Westfaelische Wilhelms-University Muenster, Institute for Experimental Epilepsy Research, Huefferstr. 68, 48149 Muenster, Germany, and the § Westfaelische Wilhelms-University Muenster, Institute of Physiology I, Robert-Koch-Str. 27a, 48149 Muenster, Germany Two major K channels are expressed in T cells, (i) the voltage- dependent K V 1.3 channel and (ii) the Ca 2 -activated K chan- nel KCa 3.1 (IKCa channel). Both critically influence T cell effec- tor functions in vitro and animal models in vivo. Here we identify and characterize TWIK-related acid-sensitive potas- sium channel 1 (TASK1) and TASK3 as an important third K conductance on T lymphocytes. T lymphocytes constitutively express TASK1 and -3 protein. Application of semi-selective TASK blockers resulted in a significant reduction of cytokine production and cell proliferation. Interference with TASK channels on CD3 T cells revealed a dose-dependent reduction (40%) of an outward current in patch clamp recordings indica- tive of TASK channels, a finding confirmed by computational mod- eling. In vivo relevance of our findings was addressed in an experi- mental model of multiple sclerosis, adoptive transfer experimental autoimmune encephalomyelitis. Pre-treatment of myelin basic protein-specific encephalitogenic T lymphocytes with TASK mod- ulators was associated with significant amelioration of the disease course in Lewis rats. These data introduce K 2 P channels as novel potassium conductance on T lymphocytes critically influencing T cell effector function and identify a possible molecular target for immunomodulation in T cell-mediated autoimmune disorders. The last decade has revealed much knowledge about the intracellular events accompanied by T lymphocyte activation following recognition of antigens bound to major histocompat- ibility complexes. K selective ion channels in T cells and their role in immune responses have been discussed for decades, since the discovery that non-selective K blockers could inhibit T cell proliferation in vitro (1–3). The role of K channels in the activation of T cells is pivotal, because opening the channels hyperpolarizes the membrane potential, which in turn increases the influx of Ca 2 via Ca 2 release-activated Ca 2 channels (CRAC) 4 (4). Signaling cascades after T cell receptor stimulation involve phospholipase C- (5)-mediated cleavage of phosphatidylinositol 4,5-bisphosphate into diacylglycerol (DAG) and inositol 1,4,5-trisphosphate, which results in a tran- sient elevation of intracellular calcium concentration ([Ca 2 ] i ) triggered by inositol 1,4,5-trisphosphate binding to inositol 1,4,5-trisphosphate receptors (5). Intracellular Ca 2 release triggers activation of CRAC chan- nels resulting in longer lasting (1 h) elevated [Ca 2 ] i manda- tory for further transcription-dependent steps of T cell activa- tion (4, 6). With the combined use of patch clamp electrophysiology and molecular biology, the voltage-depend- ent K channel 1.3 (K V 1.3 channel) and the intermediate con- ductance Ca 2 -activated channel (IKCa channel; IK Ca 1 or K Ca 3.1; which acquires its Ca 2 dependence from constitu- tively bound calmodulin) were found to be the dominating K channel types expressed in T cells (7–10). K V 1.3 is activated by membrane depolarization, whereas K Ca 3.1 channel opening is triggered by a rise in Ca 2 ions (5). Selective blockade of K V 1.3 leads to suppression of T cell effector function, e.g. decreased cytokine release and suppression of proliferation (11, 12). Underlining this finding, in vivo blockade of K V 1.3 has been shown to mediate beneficial effects in experimental autoim- mune encephalomyelitis (EAE), a rodent model for multiple scle- rosis (12, 13). The importance of K channels has been further validated by research on human T lymphocytes, which revealed a differential expression patterns of K V 1.3 and K Ca 3.1 on specific T cell subtypes (14). Interestingly, “chronically activated” T cells induced by repeated antigenic challenges of the cells selectively and stably up-regulate K V 1.3 channels (15). Furthermore, these cells could be defined as CCR 7 CD45RA effector memory T lymphocytes (16), which are implicated in different autoimmune disorders (15). In agreement with that K V 1.3 has been shown to be highly expressed in human myelin-reactive T cells of patients with multiple sclerosis (17) as well as in autoreactive T cells from patients with other autoimmune diseases such as type-1 diabetes or rheumatoid arthritis (18). Thus K channels, especially K V 1.3, have emerged as attractive targets for highly selective immunosup- pressive strategies applicable for example in the therapy of T cell- mediated autoimmune disorders (19). Two-pore-domain K channels (K 2 P, KCNK), often de- scribed as “background” or “leak” channels, play a pivotal role in * This work was supported by Interdisciplinary Clinical Research Center (IZKF) Wuerzburg N39-1 (to S. G. M. and H. W.). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1 Both authors contributed equally. 2 To whom correspondence should be addressed. Tel.: 49-931-201-23756; Fax: 49-931-201-23488; E-mail: [email protected]. 3 Submitted in partial fulfillment of a doctoral thesis, Department of Neurol- ogy, University of Wuerzburg, Wuerzburg, Germany. 4 The abbreviations used are: CRAC, calcium release-activated calcium chan- nel; K V 1.3, voltage-gated potassium channel; IKCa1, intermediate conduct- ance calcium-activated potassium channel; FACS, fluorescence-activated cell sorter; CFSE, carboxyfluorescein diacetate succinimidyl ester; EAE, experimental autoimmune encephalomyelitis; DAPI, 4,6-diamidino-2- phenylindole; IFN, interferon; IL, interleukin; PBS, phosphate-buffered saline; MBP, myelin basic protein. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 283, NO. 21, pp. 14559 –14570, May 23, 2008 © 2008 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. MAY 23, 2008 • VOLUME 283 • NUMBER 21 JOURNAL OF BIOLOGICAL CHEMISTRY 14559 by guest on September 12, 2020 http://www.jbc.org/ Downloaded from

Upload: others

Post on 20-Jul-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

TWIK-related Acid-sensitive K� Channel 1 (TASK1) and TASK3Critically Influence T Lymphocyte Effector Functions*

Received for publication, January 24, 2008, and in revised form, March 6, 2008 Published, JBC Papers in Press, March 28, 2008, DOI 10.1074/jbc.M800637200

Sven G. Meuth‡1,2, Stefan Bittner‡1,3, Patrick Meuth§, Ole J. Simon‡, Thomas Budde¶, and Heinz Wiendl‡

From the ‡Department of Neurology, University of Wuerzburg, Josef-Schneider-Str. 11, 97080 Wuerzburg, the ¶WestfaelischeWilhelms-University Muenster, Institute for Experimental Epilepsy Research, Huefferstr. 68, 48149 Muenster, Germany, and the§Westfaelische Wilhelms-University Muenster, Institute of Physiology I, Robert-Koch-Str. 27a, 48149 Muenster, Germany

TwomajorK�channelsareexpressed inTcells, (i) thevoltage-dependent KV1.3 channel and (ii) the Ca2�-activated K� chan-nelKCa3.1 (IKCa channel). Both critically influenceTcell effec-tor functions in vitro and animal models in vivo. Here weidentify and characterize TWIK-related acid-sensitive potas-sium channel 1 (TASK1) and TASK3 as an important third K�

conductance on T lymphocytes. T lymphocytes constitutivelyexpress TASK1 and -3 protein. Application of semi-selectiveTASK blockers resulted in a significant reduction of cytokineproduction and cell proliferation. Interference with TASKchannels on CD3� T cells revealed a dose-dependent reduction(�40%) of an outward current in patch clamp recordings indica-tiveofTASKchannels,a findingconfirmedbycomputationalmod-eling. In vivo relevance of our findings was addressed in an experi-mentalmodel ofmultiple sclerosis, adoptive transfer experimentalautoimmune encephalomyelitis. Pre-treatment of myelin basicprotein-specific encephalitogenicT lymphocyteswithTASKmod-ulators was associated with significant amelioration of the diseasecourse in Lewis rats. These data introduce K2P channels as novelpotassium conductance on T lymphocytes critically influencing Tcell effector function and identify a possible molecular target forimmunomodulation in T cell-mediated autoimmune disorders.

The last decade has revealed much knowledge about theintracellular events accompanied by T lymphocyte activationfollowing recognition of antigens bound tomajor histocompat-ibility complexes. K� selective ion channels in T cells and theirrole in immune responses have been discussed for decades,since the discovery that non-selective K� blockers could inhibitT cell proliferation in vitro (1–3). The role of K� channels in theactivation of T cells is pivotal, because opening the channelshyperpolarizes the membrane potential, which in turnincreases the influx of Ca2� via Ca2� release-activated Ca2�

channels (CRAC)4 (4). Signaling cascades after T cell receptor

stimulation involve phospholipase C-� (5)-mediated cleavageof phosphatidylinositol 4,5-bisphosphate into diacylglycerol(DAG) and inositol 1,4,5-trisphosphate, which results in a tran-sient elevation of intracellular calcium concentration ([Ca2�]i)triggered by inositol 1,4,5-trisphosphate binding to inositol1,4,5-trisphosphate receptors (5).Intracellular Ca2� release triggers activation of CRAC chan-

nels resulting in longer lasting (�1 h) elevated [Ca2�]i manda-tory for further transcription-dependent steps of T cell activa-tion (4, 6). With the combined use of patch clampelectrophysiology and molecular biology, the voltage-depend-ent K� channel 1.3 (KV1.3 channel) and the intermediate con-ductance Ca2�-activated channel (IKCa channel; IKCa1 orKCa3.1; which acquires its Ca2� dependence from constitu-tively bound calmodulin) were found to be the dominating K�

channel types expressed in T cells (7–10). KV1.3 is activated bymembrane depolarization, whereas KCa3.1 channel opening istriggered by a rise in Ca2� ions (5). Selective blockade of KV1.3leads to suppression of T cell effector function, e.g. decreasedcytokine release and suppression of proliferation (11, 12).Underlining this finding, in vivo blockade of KV1.3 has beenshown to mediate beneficial effects in experimental autoim-mune encephalomyelitis (EAE), a rodent model for multiple scle-rosis (12, 13). The importance of K� channels has been furthervalidated by research on human T lymphocytes, which revealed adifferential expression patterns of KV1.3 and KCa3.1 on specific Tcell subtypes (14). Interestingly, “chronically activated” T cellsinduced by repeated antigenic challenges of the cells selectivelyand stably up-regulate KV1.3 channels (15). Furthermore, thesecells could be defined as CCR7�CD45RA� effector memory Tlymphocytes (16), which are implicated in different autoimmunedisorders (15). In agreementwith thatKV1.3 has been shown to behighly expressed in humanmyelin-reactiveT cells of patientswithmultiple sclerosis (17) as well as in autoreactive T cells frompatients with other autoimmune diseases such as type-1 diabetesor rheumatoid arthritis (18). Thus K� channels, especially KV1.3,haveemergedasattractive targets forhighly selective immunosup-pressive strategies applicable for example in the therapy of T cell-mediated autoimmune disorders (19).Two-pore-domain K� channels (K2P, KCNK), often de-

scribed as “background” or “leak” channels, play a pivotal role in

* This work was supported by Interdisciplinary Clinical Research Center (IZKF)Wuerzburg N39-1 (to S. G. M. and H. W.). The costs of publication of thisarticle were defrayed in part by the payment of page charges. This articlemust therefore be hereby marked “advertisement” in accordance with 18U.S.C. Section 1734 solely to indicate this fact.

1 Both authors contributed equally.2 To whom correspondence should be addressed. Tel.: 49-931-201-23756;

Fax: 49-931-201-23488; E-mail: [email protected] Submitted in partial fulfillment of a doctoral thesis, Department of Neurol-

ogy, University of Wuerzburg, Wuerzburg, Germany.4 The abbreviations used are: CRAC, calcium release-activated calcium chan-

nel; KV1.3, voltage-gated potassium channel; IKCa1, intermediate conduct-

ance calcium-activated potassium channel; FACS, fluorescence-activatedcell sorter; CFSE, carboxyfluorescein diacetate succinimidyl ester; EAE,experimental autoimmune encephalomyelitis; DAPI, 4�,6-diamidino-2-phenylindole; IFN, interferon; IL, interleukin; PBS, phosphate-bufferedsaline; MBP, myelin basic protein.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 283, NO. 21, pp. 14559 –14570, May 23, 2008© 2008 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

MAY 23, 2008 • VOLUME 283 • NUMBER 21 JOURNAL OF BIOLOGICAL CHEMISTRY 14559

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

setting the resting membrane potential and modulating neuro-nal excitability (20–23). K2P channels consist of four trans-membrane domains arranged in tandem building two pores.Importantly, their action is mostly time and voltage independ-ent (21). TASK1 andTASK3 channels, two functionalmembersof the K2P channel family, can be regulated by a diversity ofstimuli (extracellular acidification, Gq proteins, and muscarine(20, 22–24)), and exhibit insensitivity to “classical” potassiumchannel blockers (e.g. tetraethylammonium, 4-aminopyridine).Based on their specific electrophysiological properties, phar-macological profile, and functional impact on the restingmem-brane potential they might represent a relevant ionic conduct-ance influencing basic T cell function.We therefore questionedwhether K2P channels are expressed on T lymphocytes anddissect their functional role on the level of electrophysiologicalproperties and T cell effector function, both in vitro and in ananimal model of multiple sclerosis in vivo.

EXPERIMENTAL PROCEDURES

Materials and Reagents—Charybdotoxin, bupivacaine,spermine, and ruthenium red (diluted in H2O; Sigma), Psora-4(DMSO; Carl Roth GmbH, Germany), and anandamide (EtOH;Tocris, Germany) were frozen as aliquots for further use. CD3/CD28 dynabeads for cell stimulation were obtained fromDynalBiotech (Karlsruhe, Germany). Annexin-FLUOS (Roche), pro-pidium iodide (Calbiochem, Darmstadt, Germany), DAPI(Merck, Darmstadt, Germany), and carboxyfluorescein diac-etate succinimidyl ester (CFSE) (Molecular Probes, Invitrogen)were used for cell labeling.Isolation and Culture of Human T Cells—Human T cells

were purified from peripheral blood samples of healthy donors(n � 49). Peripheral bloodmononuclear cells were prepared bydensity centrifugation (Lymphoprep, Axis-Shield, Oslo, Nor-way) according to the manufacturer’s instructions. This wasfollowed by magnetic cell sorting (MACS�, CD3 Microbeads,Miltenyi Biotec, Karlsruhe, Germany) for CD3. Purity of CD3�

T lymphocytes was �98% as assessed by flow cytometry. Cellswere maintained in RPMI 1640 containing 10% human AB-se-rum, 25 mM HEPES, 1% glutamine, and 1% antibiotics.Assessment of T Cell Function, Pharmacological Blockades—

4 � 106 freshly isolated CD3� T lymphocytes were seeded in 1ml of T cell medium per well. CD3/CD28 dynabeads (DynalBiotech) were added at a T cell to bead ratio of 1:1. Channelblockers in different concentrations were applied in parallel tobead stimulation (KV1.3 blockers, 10 and 100 nM charybdo-toxin, and 1, 10, and 100 nM Psora-4; TASK1/3 blocker, 30, 100,and 250 �M bupivacaine; TASK1 inhibitor, 3, 30, and 100 �Manandamide; TASK3 blockers, 50 �M, 500 �M, and 1 mMspermine, and 100 nM, 1�M, 10�M ruthenium red). The solventsolution in the final experimental solution did not exceed 1%.Application of the solvent alone (1%) did not influence the ana-lyzed parameters. Stimulations were all done in duplicates.After 24 h incubation at 37 °C and 5% CO2, cells were centri-fuged and subjected to further analysis by flow cytometry(stainings for annexin V and propidium iodide). In parallel,supernatants were assessed for IFN� or IL2 protein levels byenzyme-linked immunosorbent assay (R&D Systems, Wiesba-den, Germany) according to the manufacturer’s instructions.

Flow Cytometry for TASK Channel Expression on TLymphocytes—Flow cytometry acquisition was done by stand-ard methods. For antibody staining cells were resuspended inFACS� buffer (PBS containing 0.1% bovine serum albumin and0.1% NaN3), directly labeled antibodies were added. Forannexin V/propidium iodide assays, cells were resuspended inannexin binding buffer (10 mM HEPES, 150 mM NaCl, 5 mMKCl, 1 mM MgCl2, and 0.18 mM CaCl2) and stained withannexin-FLUOS (Roche) and propidium iodide (Calbiochem).For evaluation of TASK1/3 antibody specificity the followingantibodies were used: rabbit anti-TASK1 (number P0981,Sigma), goat anti-TASK1 (number SC-32067), rabbit anti-TASK3 (number AB5721, Chemicon, Ochsenhausen, Ger-many), and goat anti-TASK3 (number SC-11320, Santa Cruz,Santa Cruz, CA) followed by the appropriate secondary anti-bodies (goat anti-rabbit fluorescein isothiocyanate or donkeyanti-goat Cy3; Dianova, Hamburg, Germany) using standardprotocols for extracellular (see above) and intracellular staining(permeabilization buffer and fixation buffer from ebioscience,San Diego, CA). Data acquisition was done using a FACS Cali-bur system (BD Biosciences). Results were analyzed usingCellQuest Pro Software (BD Biosciences).T Cell Proliferation Assay in the Presence and Absence of

TASK Channel Modulators—Human T cells were labeled with4 �M CFSE (Molecular Probes) for 10 min in the dark. RPMIcontaining 15% fetal calf serum was added for 20 min to stoplabeling and was followed by three washing steps with RPMI,10% fetal calf serum. CFSE-labeled cells were suspended in T cellmedium and 1 � 105 cells per well were cultured at 37 °C and 5%CO2 in the presence of CD3/CD28 beads and ion channel modu-lators as described above. After 3 days of proliferation CFSE-la-beled cells were washed and resuspended in FACS buffer. Prolif-eration assays were performed in duplicate and analyzed by flowcytometry calculating the responder frequency (dividing thenum-ber of dividing cells by total number of cells).Immunocytochemistry of Human and Rat T Cells—Immuno-

cytochemical stainings were performed on human T cells andrat myelin basic protein (MBP)-specific T cells. Cells wereplaced on coverslips coated with poly-L-lysine (Sigma) andfixed with 4% paraformaldehyde. Subsequently, cells wereblocked with PBS containing 10% horse serum (PAA Laborato-ries, Colbe, Germany), 2% bovine serum albumin, and 0.3%Tri-ton X-100 overnight. Next, the primary antibodies (rabbitanti-TASK1, Sigma; rabbit anti-TASK3, Chemicon; rabbit anti-KV1.3, Chemicon) were added and incubated for 1 h. Cells werewashed with PBS containing 0.3% Triton X-100 and incubatedwith secondary antibodies (Cy3-conjugated rabbit anti-goat,1:100, Dianova) for another hour. Counterstaining of cell nucleiwas performed using DAPI (0.5 �g/ml, Merck). Pictures werecollected by immunofluorescence microscopy (Axiophot,Zeiss, Jena, Germany). Negative controls without the primaryantibody revealed no positive signals (data not shown).Western Blot of T Cell Lysates—Whole cell lysates from iso-

lated purified T cells were used for Western blot analysis. Inbrief, cells were washed with ice-cold PBS, resuspended in lysisbuffer (PBS containing 1% Triton X-100 and protease inhibitormixture (Roche Diagnostics, Mannheim, Germany), and solu-bilized by sonification on ice. Cell lysates were centrifuged and

TASK on T cells

14560 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 283 • NUMBER 21 • MAY 23, 2008

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

protein content in the clarified supernatant was measured byBradford reaction. Samples (50 �g/lane for TASK1 and 100�g/lane for TASK3) were subjected to 10% SDS-PAGE, fol-lowed by transfer to nitrocellulose membranes. Whole mousebrain (C57/Bl6) was used as a positive control for TASK chan-nel expression (23). Protein transfer was visualized by PonceauS staining and membranes were then blocked with PBS con-taining 0.05%Tween 20 and 5% drymilk. Themembranes wereprobed with rabbit anti-TASK1 (raised against the C-terminalpart of the channel, 1:200; Sigma) or rabbit anti-TASK3 (poly-clonal antibody against the C-terminal part of TASK3, 1:200;Chemicon), respectively. The secondary antibody was horse-radish peroxidase-conjugated donkey anti-rabbit (1:3,000;Amersham Biosciences). The antibody reaction was detectedby enhanced chemiluminescence reaction (ECL, AmershamBiosciences).Electrophysiological Recording of Whole Cell Outward Cur-

rents in T Cells—All experiments were conducted in whole cellconfiguration of the patch clamp technique. Individual humanT lymphocytes were visually identified by infrared differentialinterference contrast video microscopy (25). Starting from aholding potential of �80 mV, membrane currents wererecorded with pipettes pulled from borosilicate glass(GC150TF-10, Clark Electromedical Instruments, Kent, UK),connected to an EPC-10 amplifier (HEKA Elektronic, Lampre-cht, Germany), and filled with (inmM): K-gluconate, 95; K3-cit-rate, 20; NaCl, 10; HEPES, 10; MgCl2, 1; CaCl2, 0.5; 1,2-bis(2-aminophenoxy)ethane-N,N,N�,N�-tetraacetic acid, 1;Mg-ATP,3; Na-GTP, 0.5. The internal solution was set to a pH of 7.25with KOH and an osmolarity of 295 mOsm/kg. Extracellularsolution (in mM): NaCl, 120; KCl, 2.5; NaH2PO4, 1.25; HEPES,30; MgSO4, 2; CaCl2, 2; dextrose, 10; pH 7.2, was adjusted withNaOH and osmolarity was set to 305 mOsm/kg. Outward cur-rents were elicited by repeated 500-ms pulses from �80 to 40mV, applied at 30-s intervals. Typical electrode resistance was3–6 megohms with an access resistance of 6–15 megohms.Series resistance compensation ofmore than 40%was routinelyused. Voltage clamp experiments were governed by Pulse soft-ware (HEKA Elektronic) operating on an IBM compatible PC.A liquid junction potential of 6 � 2 mV (n � 6) was measuredand taken into account according to Neher (26).Adoptive Transfer Experimental Autoimmune Encephalo-

myelitis—Female Lewis rats (150–160 g) were kept at stand-ard conditions with free access to food and water. All animalexperiments were approved by local authorities and con-ducted according to the German law of animal protection.Adoptive transfer experimental allergic encephalomyelitiswas induced using a specific rat MBP T cell line (MBP-TC),which was generated as described previously (27). TheMBP-TCs used for this experiment were freshly restimulated for 3days with antigen presenting cells and guinea pig MBP protein(in one Petri dish: 3 � 106 MBP-TCs � 150 � 106 antigenpresenting cells � guinea pig MBP 10 �g/ml in 10 ml ofrestimulation medium, which consists of RPMI 1640 contain-ing 1% glutamine, 1% antibiotics, 1% rat serum, and 0.4%�-mercaptoethanol). After 24 and 48 h cells were split and fedRPMI 1640 containing 1% glutamine, 1% antibiotics, 5% fetalcalf serum, and 7.5% supernatants of concanavalin A-activated

spleen cells. 24 h before cell transfer, channel blockers (10 nMPsora-4, 30 �M anandamide, and 100 nM charybdotoxin) wereadded directly into the cell cultures. After separation from theantigen presenting cells by Ficoll gradients, 6 � 106 MBP-TCswere transferred into four groups of Lewis rats intravenously(each n� 8–9) or plated for another 24 h for evaluation of IFN�production. The animals were weighed and scored every day bya blinded investigator. Disease severity was assessed following awell established 6-grade scoring system: 0 � healthy; 1 �weight loss, limp tip of tail; 2 � limp tail, mild paresis; 3 �moderate paraparesis, ataxia; 4 � tetraparesis; 5 � moribund;6 � death (28).Numeric Model of T Cell Outward Currents—A single com-

partment T cell model including currents IKv1.3, ITASK, and ICwas developed within the NEURON Simulation Environment(29). The length and diameter of the compartment were set to10 �m each, resulting in a total membrane area of 314 �m2. Byassuming the following ion distribution (mM): K�

in � 135,K�

out � 3.5, Ca2�in � 6.5 � 10�4 (30), Ca2�

out � 0.13, wederived a potassium reversal potential (Ek) of about �94 mVand a calcium reversal potential (ECa) of about 98mV followingthe Nernst equation. A specific membrane capacitance of 1�F/cm2 was applied and all simulations were executed at 25 °C.

The delayed rectifier current IKv1.3 was realized by adaptingthe membrane mechanism IKD from a Purkinje cell model (31).Shifting the half-maximum activation and inactivation to�28.8 and �57.8 mV, respectively, led to a current thatmatches the slowly inactivating current component seen inhuman T cells (32, 33). The maximum specific conductancegkbar was set to 0.0023 S/cm2 to reach a maximum current ofabout 200 pA at a membrane potential of �40 mV. The Ikcurrent amplitude was calculated according to the followingequation: Ik � gkbar � m � h � (VM � Ek);m, activation statevariable; h, inactivation state variable; andVM,membrane volt-age. Including the potassium leak current ITASK into the modelwas accomplished by using the corresponding membranemechanism as described earlier (34). The described kinetic ofthat current thereby remained unaffected, whereas the maxi-mum specific conductance was adapted to reach a maximumcurrent of about 200 pA at a membrane potential of �40 mV.The calcium-dependent potassium current IC originates fromArthur Houweling’s NEURON demo. Kinetics of the currentwere described previously (35) and remained unchangedduring all simulations. Setting the maximum specific con-ductance gkbar to 0.00034 S/cm2 resulted in an approximate115 pA at a membrane potential of �40 mV. The followingequation describes this non-inactivating current: Ik �gkbar � m � (VM � Ek), where Ik, current amplitude; m,activation state variable; and VM, membrane voltage.Statistical Analysis—All results are presented as mean � S.E.

Statistical analysis was performed using the Student’s t test modi-fied for small samples as described previously (36). Statistical sig-nificancewassetatp0.05,which is indicatedbydoubleasterisks.

RESULTS

TASK1 and TASK3 Channels Are Expressed on Human TLymphocytes—In a first set of experiments purified humanCD3� T lymphocytes (n � 6 donors) were assessed by immu-

TASK on T cells

MAY 23, 2008 • VOLUME 283 • NUMBER 21 JOURNAL OF BIOLOGICAL CHEMISTRY 14561

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

nocytochemistry for the expression of K2P channels TASK1and TASK3. Voltage-gated KV1.3 channel was stained as a pos-itive control (18). Immunoreactivity for TASK1, TASK3, andKV1.3 was clearly detectable (Fig. 1, A–C). Counterstaining ofcell nuclei with DAPI indicated a membrane-bound distribu-tion pattern for all channels, whereas incubation with the sec-ondary antibody alone showed no signals (data not shown).Protein expression of TASK1 and TASK3 by humanT cells wascorroborated by Western blot analysis (n � 6 donors). Theband recognized by the anti-TASK1 antibody was �50 kDa(Fig. 1D, lanes 4–6) as published earlier (37). In human lympho-

cytes as well as in our positive control (wholemouse brain, lane3) a second band was visible at 65 kDa (Fig. 1D, second arrow),not visible in specificity controls (Fig. 1D, lanes 1 and 2). Usingan anti-TASK3 antibody a distinct band at 60 kDa was visible,both in the mouse brain and human T lymphocytes (Fig. 1E,lanes 2–5). To verify antibody specificity, different antibodies forTASK1 andTASK3were also assessed by flow cytometry. Anti-bodies fromSigma (TASK1) and SantaCruz (TASK3) recogniz-ing intracellular epitopes of these channels showed specificbinding only in intracellular FACS staining with permeabilizedcells (Fig. 1F, right side) and not in extracellular staining proto-

FIGURE 1. Expression of K� channels KV1.3, TASK1, and TASK3 on human T lymphocytes. A, immunocytochemical staining of human CD3� T lymphocyteswith DAPI (left panel), anti-KV1.3 (middle panel), and overlay (right panel). B and C, antibodies raised against TASK1 (B, middle panel) and TASK3 (C, middle panel)and corresponding DAPI counterstaining (B and C, left panels) indicates a channel expression on human T lymphocytes (B and C, right panels). Scale barsrepresents 50 �m (upper left) and counts for all images (enlarged details scale bar, 10 �m). D, Western blot analysis using a TASK1-specific antibody revealedtwo distinct signals at 50 and 65 kDa (3). Left lane indicates marker bands. Lanes 1, negative control; 2, albumin blotting (no band); 3, whole mouse brain; and4 – 6, Lysates of CD3-positive isolated T cells. E, Western blot of TASK3 protein indicates a clear signal at 60 kDa (3) using CD3-positive isolated T cells of healthyhumans (lanes 3–5). Marker bands are indicated on the left lane. Lanes 1, negative control; 2, whole mouse brain; and 3–5, three lysates of CD3-positive isolatedT cells from 3 independent healthy donors. F, flow cytometry for TASK1 (upper panel) and TASK3 (lower panel). Fluorescence intensity is plotted against eventcounts. Dotted lines, unstained controls; thin black lines, staining with secondary antibodies alone; bold black lines, staining with TASK1/3 antibodies. Arrows (2)indicate overlapping signals for thin and bold black lines. One of three representative experiments is shown.

TASK on T cells

14562 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 283 • NUMBER 21 • MAY 23, 2008

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

cols (Fig. 1F, left side). The same specificity was found for ananti-TASK1 antibody (intracellular target) by Santa Cruz,whereas anti-TASK3 (extracellular; Chemicon) revealed posi-

tive signals in intracellular andextracellular staining protocols(data not shown). Taken together,these results point to expression ofK2P channels TASK1 and -3 onhuman CD3� T lymphocytes.TASK1 and TASK3 Modulate T

Cell Effector Function: Pharmaco-logical Blockade Inhibits IFN�Secretion, IL2 Secretion, and T CellProliferation—To correlate K2Pchannel expression to T cell effectorfunctions we next studied the influ-ence of pharmacological blockingreagents for K� channels on cyto-kine production and T cell prolifer-ation. Human CD3� T cells werestimulated with CD3/CD28 beads.Levels of IFN� and IL2 secreted inthe supernatants were assessed inresponse to application of differention channelmodulators. To excludeunspecific effects of the compoundson cell viability, each inhibitor wastitrated and tested by annexinV/propidium iodide staining to esti-mate T cell apoptosis and necrosis.The gating procedure, results undercontrol conditions, and after treat-ment with H2O2 (100 �M, positivecontrol for cell death) are depictedin Fig. 2A. Charybdotoxin (C, 10 nM,100 nM) and Psora-4 (P, 1, 10, and100 nM), both inhibitors of KV1.3channels, the TASK channel inhibi-tor bupivacaine (B, 30, 100, and 250�M), and anandamide (A, 3, 30, and100 �M) as well as the TASK3 mod-ulating compounds spermine (S, 50�M, 500 �M, and 1 mM) and ruthe-nium red (R, 100 nM, 1 �M, and 10�M) had no significant direct effecton cell survival (Fig. 2, B–D, upperpanel) as compared with controlconditions (con). Charybdotoxinand Psora-4, both modulators ofKV1.3 were used as positive controls(38). Application of 100 nM charyb-dotoxin significantly reduced IFN�production to 56 � 3% (n � 5, p �0.0002; 10 nM: no effect, n � 5, p �0.76; Fig. 2B;C1,C2). KV1.3 channelmodulation by Psora-4 significantlylowered IFN� amounts in the super-natant at all concentrations used

(Fig. 2B and Table 1). TASK channel modulation using bupiva-caine (23) significantly decreased IFN� levels at concentrations�100�M (Fig. 2C andTable 1). TASK channel inhibition by the

FIGURE 2. Production of interferon � and IL2 by human T lymphocytes is reduced in the presence ofion channel modulators against K� channels KV1.3, TASK1, and TASK3. A, cell viability assessment inthe presence and absence of K� channel modulators was performed by flow cytometry. Gating procedures(side and forward scatter), results under control conditions, and after treatment with H2O2 (100 �M) aredepicted. B, upper panel, superimposed images exemplify flow cytometry analysis for apoptosis and necrosisunder control conditions (con) and in the presence of the different channel modulators charybdotoxin (100 nM,C) and Psora-4 (100 nM, P). Lower panel, interferon � production of isolated human T lymphocytes after stimu-lation with CD3/CD28 beads under control conditions (positive control; black column, �) and in the presence ofthe KV1.3 channel inhibitors charybdotoxin (C1, 10 nM; C2, 100 nM) and Psora-4 (P1, 1 nM; P2, 10 nM; P3, 100 nM).C, IFN� production after application of bupivacaine (B1, 30 �M; B2, 100 �M; B3, 250 �M) and anandamide (A1, 3�M; A2, 30 �M; A3, 100 �M). D, TASK3 specific inhibition by spermine (S1, 50 �M; S2, 500 �M; S3, 1 mM) andruthenium red (R1, 100 nM; R2, 1 �M; R3, 10 �M). E, blockade of KV1.3 and TASK channels using selectedconcentrations leads to a similar effect on IL-2 production. **, p 0.05.

TASK on T cells

MAY 23, 2008 • VOLUME 283 • NUMBER 21 JOURNAL OF BIOLOGICAL CHEMISTRY 14563

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

endogenous cannabinoide anandamide significantly reducedIFN� secretion in a dose-dependent manner (e.g. 30 �M: 68 �8%, n � 5, p � 0.02; Fig. 2C and Table 1). Similarly, the TASK3channel modulator spermine suppressed IFN� secretion atconcentrations�500�M (Fig. 2D andTable 1). Ruthenium red,a second inhibitor of TASK3, similarly suppressed IFN� pro-duction by human T cells (e.g. 100 nM: 47 � 6%, n � 5, p �0.0007; Fig. 2D and Table 1). Analysis for IL2, a second impor-tant cytokine for T lymphocyte effector functions, revealedsimilar results for KV1.3 and TASK blockade (C1, 56 � 7%, n �5, p� 0.03;P2, 74� 3%, n� 5, p� 0.05;A2, 45� 5%, n� 5, p�0.001; R2, 53 � 7%, n � 5, p � 0.02; Fig. 2E). In addition, weassessed the effect of TASKmodulation on T cell proliferation.CFSE dilution was measured after 3 days of CD3/CD28 stimu-lation (positive control, normalized to 100%) and comparedwith control values (Fig. 3A and Table 2). Unstimulated T cellsdisplayed a responder frequency of 6� 1% (Table 2). In accord-ance with the literature, KV1.3 modulation (charybdotoxin,

Psora-4) significantly reduced responder frequency after beadstimulation (n � 5 each; Fig. 3B, Table 2). TASK channel mod-ulation by bupivacaine (n� 5; Table 2), anandamide (n� 5; Fig.3C, Table 2), spermine (TASK3, n� 5; Table 2), and rutheniumred (TASK3, n � 5; Fig. 3D and Table 2) significantly sup-pressed T cell proliferation (30–70%; Table 2).Electrophysiological Recordings Reveal the Contribution of

TASK to the PotassiumOutwardCurrent inHumanTCells—Inthe next set of experiments whole cell patch clamp recordingsof purified CD3� human T lymphocytes were used to evaluatethe contribution of TASK channels to the potassium outwardcurrent of T cells (n� 31 donors;multiple single cell recordingsof each donor). Stepping the membrane potential from �80 to�40 mV (Fig. 4A, left panel, inset) under voltage-clamp condi-tions evoked an outward current of 424 pA (range: 213–944pA). Application of the voltage protocol at 30-s intervals indi-cated a stable outward current over time and current run-downcould by analyzed as 6 � 2% (Fig. 4, A, left panel; C, left andmiddle panel, n � 6). Calculation of the underlying time con-stant (�) under control conditions revealed a single exponentialdecay with 195 � 15 ms (Fig. 4, A, left panel; C, right panel andinset; n � 6). However, addition of the well established KV1.3channel inhibitor Psora-4 (100 nM) immediately reduced theoutward current by 54.8� 8.8% (Fig. 4A, right panel, gray trace;n � 4, p � 0.003) although the time constant was not signifi-cantly altered (�: 217 � 17.3 ms). In contrast, applying theTASK-specific modulator anandamide resulted in a rapidreduction of the K� outward current (30 �M, 42 � 10%, n � 4,p� 0.01; 100�M, 44� 8%, n� 4, p� 0.0017; Fig. 4,B andC, leftand middle panels) and an accelerated time constant (30 �M,� � 80� 17ms; n� 4, p� 0.0002; 100�M, � � 69� 10ms; n�4, p � 0.00001; Fig. 4C, right panel). These results for the firsttime demonstrate a contribution of currents throughK2P chan-nels to outward currents in human T cells. Next, blocker-sen-sitive currents were analyzed after graphical subtraction of cur-rents in the presence and absence of channel modulators.Expectedly, the Psora-4-sensitive current component (the cur-rent after application of Psora-4 was subtracted from the con-trol current, Fig. 4A, right panel, black trace minus gray trace)revealed a rapid current onset upon stimulation and a slowinactivation kinetic indicative of the delayed rectifying currentKV1.3 (Fig. 4D, left panel). However, the anandamide-sensitivecurrent component (the current after application of anandam-ide was subtracted from the control current, Fig. 4B, left panel,

FIGURE 3. Modulation of TASK inhibits proliferation of human T lympho-cytes. Human CD3 T lymphocytes were labeled with CFSE and cultured in thepresence of CD3/CD28 beads (A) and various K� channel modulators (B–D).Application of the potassium channel modulators Psora-4 (P, 10 nM, n � 5),anandamide (A, 30 �M, n � 5), and ruthenium red (R, 100 nM, n � 5) indicateda marked reduction of T cell proliferation. Exemplary histograms are shown.See corresponding Table 2.

TABLE 1IFN� production in the absence and presence of K� channelmodulators

Substance Abbreviation Concentration Result n p valuePsora-4 P1 1 nM 64 � 8% 5 0.01

P2 10 nM 61 � 8% 5 0.007P3 100 nM 54 � 12% 5 0.02

Bupivacaine B1 30 �M 96 � 4% 5 0.44B2 100 �M 74 � 6% 5 0.01B3 250 �M 59 � 12% 5 0.00001

Anandamide A1 3 �M 81 � 4% 5 0.01A2 30 �M 68 � 8% 5 0.02A3 100 �M 26 � 2% 5 0.00001

Spermine S1 50 �M 82 � 8% 5 0.1S2 500 �M 62 � 4% 5 0.001S3 1 mM 55 � 5% 5 0.0007

Ruthenium red R1 100 nM 47 � 6% 5 0.0007R2 1 �M 40 � 8% 5 0.002R3 10 �M 33 � 6% 5 0.0005

TABLE 2The table summarizes all investigated compounds andconcentrations of the inhibitors used in this study concerninghuman T-cell proliferation

Responder frequency S.E. p value%

Unstimulated 6 1 0.00001CD3/CD28 stimulated 100 0 0Charybdotoxin, 100 nM 61 6 0.002Psora-4, 10 nM 27 7 0.004Bupivacaine, 250 �M 52 8 0.004Anandamide, 30 �M 70 9 0.02Spermine, 500 �M 67 12 0.046Spermine, 1 mM 60 11 0.02Ruthenium red, 1 nM 98 4 0.46Ruthenium red, 10 nM 76 4 0.003Ruthenium red, 0.1 �M 28 8 0.0007

TASK on T cells

14564 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 283 • NUMBER 21 • MAY 23, 2008

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

black traceminus gray trace) displayed a fast current onset withnearly no inactivation over the stimulation protocol, a typicalfeature of voltage-independent TASK channels (Fig. 4D, right

panel). These findings were furthercorroborated by the electrophysi-ological fingerprint of charybdo-toxin (50 nM), a second blocker ofKV1.3 (and KCa3.1), on human Tlymphocytes (64.5 � 6.4% reduc-tion, n� 4, p� 0.0002; Fig. 5A, insetand right panel). The charybdo-toxin-resistant currents (Fig. 5A,left panel) strongly resemble theproperties of pure TASK currents,which provides further support forthe coexistence of voltage-de-pendent and leak potassium cur-rents on T cells. TASK3 modula-tion by ruthenium red (1 and 10�M) induced a significant currentreduction of 40.9 � 8.5 and 52.6 �4%, respectively (n � 4, p � 0.04/0.001; Fig. 5B). Taken together thisdata therefore clearly indicate thefunctional relevance of TASKchannels on outward currents inhuman T lymphocytes and dem-onstrate the functional coexpres-sion of KV1.3 and TASK channelson these cells.A Computational Model to Dis-

sect the Contribution of Three KChannels (KV1.3, KCa, and TASK)for K� Outward Currents—In thenext experimental step we used anumeric model of human T cells toanalyze the current components

contributing to the resting membrane potential in human Tcells. The model included the voltage-dependent KV1.3 (IKv1.3)channel (Fig. 6A), whereas inactivation and activation parame-ters were calculated according to the literature (31, 39). Basedon these settings the currents evoked by a depolarizing voltageprotocol (500ms, from�50 to�50mV, decrement 10mV; Fig.6A, inset) showed slow inactivation kinetics and characteristicsas described for the net outward current of human T cells (16,33). Next, the model was extended by the inclusion of a TASKcurrent (ITASK) as described earlier (34). TASK current aloneresulted in a nearly voltage-independent model response todepolarizing voltage steps (Fig. 6B). Finally, we supplementedthe model by including a Ca2�-dependent K� current (IC, Fig.6C) (35). Based on our pharmacological results we assumed thecontribution of all three conductances to the net outward cur-rent in human T cells and calculated 40% IKv1.3, 40% ITASK, and20% IC. The composed current evoked by a depolarizing voltagestep to �40 mV displayed typical features (e.g. slow inactiva-tion) as recorded for humanT lymphocytes (Fig. 6D, left panel).Mimicking Psora-4 actions (inhibitor of KV1.3 channels asdemonstrated by electrophysiological recordings) in the modelreduced the KV1.3 component to 0% resulting in a nearly volt-age independent current response evoked by a depolarizingvoltage step (Figs. 4A, right panel, gray trace, and 6D, middle

FIGURE 4. Whole cell patch clamp recordings of potassium outward currents in human T cells reveal apharmacological profile indicative of KV1.3 and TASK channels. A, voltage step protocols (see inset) from�80 to �40 mV (500 ms, repeated every 30 s) were used to evoke stable potassium outward currents on humanT lymphocytes (left panel). Application of Psora-4 (100 nM, inhibitor of KV1.3 channels) induced a clear currentreduction (right panel). B, the endogenous cannabinoid anandamide (30 and 100 �M) significantly reduced thecurrent. C, bar graph representation of rundown effects under control conditions (con) compared with TASK-specific inhibition by anandamide (30 �M, left panel). Amplitude of the net outward current plotted againsttime under control conditions (black squares, middle panel). Period of anandamide treatment (ana, gray circles)is indicated by a horizontal line (middle panel). Bar graph representation of time constants under control con-ditions and after application of anandamide (right panel) as indicated by mathematical single-exponentialdecay fits (right panel, inset). D, blocker-sensitive current components obtained by subtraction of currents afterapplication of an inhibitor from control values. ** represents p 0.05.

FIGURE 5. Effects of charybdotoxin and ruthenium red on the potassium out-ward currents in human T cells. A, effects of charybdotoxin (50 nM) on the netoutward current are shown in the left panel. Mean bar graph representation (inset)and amplitude versus time plot of charybdotoxin (right panel) are shown. B, ruthe-nium red (RR) applied on the T cell outward current induced a significant currentreduction at 1 and 10 �M (left panel). Amplitude of the net outward current plot-ted against time in the presence of 1 �M ruthenium red as indicated by a horizon-tal line (middle panel). Bar graph representation of ruthenium red effects on thenet outward current of human T lymphocytes compared with control values(right panel). **, p 0.05.

TASK on T cells

MAY 23, 2008 • VOLUME 283 • NUMBER 21 JOURNAL OF BIOLOGICAL CHEMISTRY 14565

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

panel). This finding further corroborates our electrophysi-ological data above, suggesting amarked contribution of TASKchannels to the outward current of human T cells. In a last stepwe modeled anandamide actions (TASK channel inhibitor asdemonstrated by electrophysiological recordings) by reducingITASK to 0%. The remaining current component representsKv1.3 channels (Figs. 4B, left panel, gray trace, and 6D, rightpanel). Recapitulating these findings from a numerical cellmodel support the results from electrophysiological recordingsshowing the functional coexpression of TASK and KV1.3 chan-nels in human T lymphocytes.Selective Blockade of T Lymphocyte TASK Channels Amelio-

rates Experimental Autoimmune Encephalomyelitis, aModel of

Multiple Sclerosis—Guided by ourdata on the functional impact ofTASK on T cell activation in vitroand our patch clamp results, wechallenged the question whetherselective modulation of T lympho-cyte TASK channels modulates Tcell-mediated inflammatory disor-ders in vivo. We therefore choseadoptive transfer experimentalautoimmune encephalomyelitis, awell established animal model ofmultiple sclerosis in Lewis rats. Toevaluate the effect of TASK modu-lation on T cells, we incubatedencephalitogenic MBP-specific Tlymphocytes with the K� channelmodulators (10 nM Psora-4, 100 nMcharybdotoxin, and 30 �M anand-amide) prior to adoptive transfer inrats. Expression of KV1.3 as well asTASK1 could be demonstrated onMBP-specific rat T lymphocytes byimmunocytochemistry, similar toour results observed in human Tlymphocytes (Fig. 7A). Intravenoustransfer of restimulated MBP-spe-cific (6 � 106 cells/animal) T cellselicited a typical EAEwith symptomonset at 2–3 days, diseasemaximumaround day 5, and recovery fromdays 6–10 (onset, 3.3 � 0.3 days,n � 8; disease maximum, score4.7 � 0.5, n � 8; decline to baseline,10.4 � 0.5 days, n � 8; Fig. 7, C–E).Incubation of encephalitogenic Tcells with the Kv1.3 blocker Psora-424 h prior to transfer led to a slightlydelayed onset of disease (not signif-icant, ns, p � 0.09), amelioration ofthe disease maximum (score 3.0 �0.6, n � 8, p � 0.03), and fasterrecovery of symptoms (p � 0.03;Fig. 7C). Different results were seenwhen using charybdotoxin that

showed no significant effect on the investigated parameters(onset, p � 0.25; disease maximum, score 4.8 � 0.8, p � 0.74,n� 8; decline to baseline, p� 0.57; Fig. 7D). Selective blockadeof TASK channels with anandamide ameliorated adoptivetransfer experimental allergic encephalomyelitis. Although dis-ease onset was delayed in the presence of anandamide (ns, p �0.39), the maximum disease score was reduced significantly(2.8 � 0.3, n � 9, p � 0.02). Furthermore, some animals recov-ered earlier (p � 0.03; Fig. 7E). In agreement with these resultsfrom EAE, MBP-specific T cells showed a significantly reducedIFN� production in vitro when pretreated with Psora-4 (0.71�0.08, p � 0.02, n � 5) and anandamide (0.66 � 0.09, p � 0.01,n � 5), whereas charybdotoxin failed to have a lasting effect on

FIGURE 6. Computational modeling of K� channel contributions to the net outward current in human Tcells. A single compartment cell model calculating the effects of KV1.3, TASK1, and 3 channels. We modeled aT cell outward current consisting of IKv1.3 (A), ITASK (B), and IC (C). A–C, left column indicates the currents evokedby depolarizing voltage steps (inset) if only a single current is included in the model. Right panel demonstratesthe activation and inactivation curve of KV1.3 (A, right column), the current-voltage relationship of TASKchannels (B, right column), and the activation curve of IC (C, right column). Analysis of the net outwardcurrent (40% IKv1.3, 40% ITASK, and 20% IC) demonstrated characteristic features comparable with therecorded outward current of human T lymphocytes (D, left panel). Different K� channel contributions tothe net outward current were modeled and revealed results comparable with the electrophysiologicalrecordings under Psora-4 (D, middle panel) and anandamide (D, right panel).

TASK on T cells

14566 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 283 • NUMBER 21 • MAY 23, 2008

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

T cells (0.84 � 0.15, p � 0.32, n � 5). This could be due todifferent binding affinities of the used substances (Fig. 7B). Ofnote, the application of channel modulators in vivo was notassociated with obvious side effects. Taken together, resultsobtained from the animal model demonstrate the pathophysi-ological relevance of TASK channel modulation in a T cell-mediated disorder.

DISCUSSION

Potassium-selective ion channels play a key role in modulat-ing effector functions of T lymphocytes. Based on convincingdata in vitro and in animal models of autoimmune disorders invivo, their selective pharmacological intervention has been pro-posed as a potential therapeutic strategy for T cell-mediatedautoimmune disorders (12, 16). Thus far, research reports havefocused on 2 major K� channels expressed in T cells, (i) thevoltage-dependent KV1.3 channel and (ii) the Ca2�-activated Kchannel KCa3.1. Pharmacological interventions of both typesinhibit T effector functions in vitro and ameliorate disease inanimal models of T cell-mediated autoimmunity in vivo (17).TASK Channel Expression on Human T Lymphocytes: Implica-

tions for T Cell Effector Functions—Our report identifies andcharacterizes the K2P channels TASK1 and -3 as a third K�

conductance relevant for T lympho-cyte activation and function. TASKchannel expression was demon-strated by immunocytochemicalstainings aswell asWestern blottingtechniques. Functionally TASKchannels contribute to the mem-brane resting potential of T cells andcritically affect T cell receptor-me-diated effector functions in vitro.This was evidenced by experimentsmeasuring cytokine secretion andcell proliferation in the presence oftwo pharmacological TASK inhibi-tors and whole cell patch clamprecordings of human CD3� T cells.Computational modeling corrobo-rated and extended electrophysi-ological dissection of the role ofTASK channels among three rele-vant K� conductances on T cells(KV1.3, KCa3.1, and TASK) andsupport a hypothetical model, howTASK channels contribute to Ca2�

entry and stabilization of the mem-brane potential. Finally, pathophys-iological relevance of our findingswas demonstrated by testing T cellselective pharmacological blockadeof TASK channels in experimentalautoimmune encephalomyelitis, ananimal model of multiple sclerosis:TASK inhibition was associatedwith slightly delayed disease onset,significant amelioration of disease

severity, and significant earlier recovery after adoptive transferof pretreated encephalitogenic cells.Role of Potassium Currents in Ca2�-dependent T Cell

Activation—Interaction of the T cell receptor-CD3 complexwith the antigen-loaded major histocompatibility complexmolecules initiates intracellular signaling via the phospholipaseC� pathway, resulting in an initial release of Ca2� from intra-cellular stores, increased intracellular Ca2�, activation of pro-tein kinase C, and augmented expression of KV1.3 and IK chan-nels in the cell membrane. Ca2� re-enters the cell throughCRAC channels, causingmembrane depolarization and furtherraising intracellular Ca2� levels. The resulting K� efflux hyper-polarizes the membrane and maximizes the driving force forcontinued Ca2� influx via CRAC channels. T cell response toantigenic stimulation is highly modulated by the shape andnature of this calcium signal indicating the importance of dif-ferences in this signaling pattern (16).The ionic conductances thus far described to be responsible

for calcium signaling in T lymphocytes are the voltage-gatedpotassium channel (KV1.3), the calcium-activated potassiumchannel (IKCa1), and CRAC (3, 7, 11, 14, 40, 41). Interestingly,a broad spectrum of antigenic and mitogenic stimuli of T cellsup-regulate expression of IK channels and also KV1.3 channels

FIGURE 7. T cell selective modulation of KV1.3 and K2P attenuates disease in an animal model of multiplesclerosis. A, expression of KV1.3 (upper row) and TASK1 (lower row) protein in rat MBP-specific T lymphocytes.Left column shows DAPI stainings, right columns the overlay. Representative stainings are shown, scale barrepresents 10 �m. B, activated MBP cells pretreated with channel modulators were incubated another 24 h andsupernatants were assessed for IFN� protein levels (un, control cells; C, charybdotoxin 100 nM; P, Psora-4 10 nM;A, anandamide 30 �M). C–E, disease score after transfer of unmanipulated MBP-specific encephalitogenic Tcells into Lewis rats is blotted as a solid line. Mean score of all animals including S.E. are shown (n � 8 –9 eachexperiment). Pretreatment of encephalitogenic T cells MBP-TC with Psora-4 (C), charybdotoxin (D), and anan-damide (E) resulted in an altered disease course. **, p 0.05.

TASK on T cells

MAY 23, 2008 • VOLUME 283 • NUMBER 21 JOURNAL OF BIOLOGICAL CHEMISTRY 14567

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 10: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

in vitro (8, 42). The increased expression of the K� channelssignificantly augments the hyperpolarizing capacity of the acti-vated T cells (compared with naıve cells), accelerates the Ca2�

influx, and secures a sustained high level of intracellular Ca2�,which is necessary for full-blown T cell proliferation and cyto-kine production. Accordingly, in the presence of K� blockers,the capability of the T cells to maintain a negative membranepotential and a long-lasting Ca2� signal is reduced, which is therationale for their putative efficacy in attenuating T cell-medi-ated immune responses.Role of K2P Channels in T Cell Physiology: Electrophysiology

and Computational Modeling—What might be the role of ournewly described K2P channels in T cells in this complex inter-play? K2P represent a unique family of potassium channels andare mainly responsible for setting the membrane potential in anumber of different cell types (20, 34, 43–46). Given the insen-sitivity of TASK channels against classical potassium channelblockers, these channels showa response against a unique panelof inhibitors including bupivacaine (23, 47–51), anandamide(35, 52), spermine (53), and ruthenium red (53, 54). Pharmaco-logical interference with TASK channels attenuates T cell prolif-eration and cytokine secretion, similar to the interference withKV1.3 usingwell established channel blocking compounds such asPsora-4 (38) and charybdotoxin (55).Notably, the inhibitory effecton T cell activity was comparable between the KV1.3 silencingsubstances Psora-4 and charybdotoxin (note here that charybdo-toxin also acts on IKCa1 channels) and the TASK channel inhibi-tors strongly suggestingan important functional impactof the leakchannels on T-cell function.Contribution of TASK currents to the net outward current of

human T lymphocytes was assessed using whole cell patchclamp recordings. A well established protocol stepping themembrane potential from �80 to �40 mV over 500 ms every30 s elicited a marked outward current with a rapid onset and aslow inactivation (single exponential decay, � � 195 ms) asdescribed earlier (16). Expectedly, application of KV1.3 channelmodulators resulted in a marked current reduction, whereascurrent kinetics remained unchanged (16). The endocannabi-noid anandamide, however, a well known inhibitor of TASKchannels (35, 52), induced a comparable inhibition of the out-ward current, but in contrast to KV1.3 blockers anandamidesignificantly altered current kinetics. Blocker-sensitive cur-rents obtained by graphical subtraction of currents after appli-cation of the inhibitor from currents under control conditionsshowed nearly no voltage dependence in the anandamide-sen-sitive component, therefore suggesting the contribution of theTASK leak current (23, 34, 35). These findings were corrobo-rated by results from a single-compartment computationalmodel including currents IKv1.3, ITASK, and IC. Furthermore,functional expression of K2P channels could be shown on iso-lated human CD3� T lymphocytes and all of the recordedCD3� T cells showed currents indicative of TASK channels. Itis currently unknown how (or if) expression of these channels isregulated (e.g. under inflammatory conditions). Furthermore,the investigation of expression levels in different T cell subtypes(naıve, Th17 encephalitogenic T cells, regulatory T cells, CD4�,CD8�, T central memory cells (Tcm), and T effector memorycells (Tem)) is warranted to determine whether TASK channels

correlate to specific T cell functions (similar toKV1.3 channels).However, based on the abundant expression of the K2P channelin a number of different cell types (20, 34, 43–46) it might bespeculated that these channels are expressed throughout differ-ent T cell subtypes and that influence of TASK on K� conduc-tances represents a “baseline” feature common to all T cells.Our findings imply the possibility that earlier results concern-ing the net outward current in human T lymphocytes could becontaminated by a TASK channel component, not known orextrapolated at that time. However, the exact role of each com-ponent contributing to the outward current inT cell can only beestimated: based on our pharmacological and modeling resultswe suggest a nearly equal contribution of currents throughTASK channels (�40%) andKV1.3 channels (�40%) to the out-ward current in human CD3� T cells, whereas other potassiumconductances (e.g. IKCa1) contribute to the remaining �20%.Taken together, our data support the following hypotheticalmodel for K2P channels in the cascade of TCR-mediated T cellactivation, as summarized in the schematic in Fig. 8.Selective Blockade of T Lymphocyte TASK Channels Amelio-

rates Experimental Autoimmune Encephalomyelitis, aModel ofMultiple Sclerosis—To prove the pathophysiological relevanceof T lymphocyte TASK channels we investigated their modula-tion in experimental autoimmune encephalomyelitis (27).Selective blockade of T lymphocyte K� channels was achievedby preincubation of encephalitogenic MBP lines prior to trans-fer with various blockers. This approach was used to dissect therelevance on T cells, which would have been contaminatedwhen applying the channel blockers systemically. In accord-ancewith the literature, KV1.3 blockade by Psora-4 amelioratedEAE: animals showed a delayed disease onset (nonsignificant),less severe disease course (significant), and a tendency toward

FIGURE 8. Hypothetical model for K2P channels in the cascade of TCR-mediated T cell activation. T cell receptor (TCR) activation is accompaniedby an increased intracellular ([Ca2�]i) Ca2� concentration. This up-regulationof Ca2� is transferred to a sustained intracellular Ca2� elevation by activationof CRAC, resulting in membrane depolarization. Furthermore, the increasedCa2� concentration leads to long lasting changes in transcription processesand T cell proliferation. To maintain the driving force for Ca2� entry throughthe plasma membrane, the Ca2�-induced depolarization is counterbalancedby potassium channels. Our data suggests that �40% of the K� outwardcurrent in human CD3� T lymphocytes is mediated by KV1.3, �20% by IKCa1,and �40% by K2P channels, a third and novel K� conductance in human Tcells. Based on their pharmacological profile (e.g. current inhibition by O2depletion or extracellular acidification) we hypothesize that TASK channelsinfluence T cell actions under (patho-)physiological conditions (adapted fromRef. 42).

TASK on T cells

14568 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 283 • NUMBER 21 • MAY 23, 2008

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 11: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

faster recovery (Fig. 7). Importantly, TASK channelmodulationvia anandamide resulted in very comparable results indicatingthat TASK-mediated effects on T lymphocytes are of patho-physiological relevance in a model of T cell-mediated autoim-munity. It is therefore tempting to speculate that attenuation ofT cell function by selective pharmacological interference withTASK channels may translate into clinical benefits in T cell-me-diated (auto)immune disorders. However, based on the broadexpression pattern of TASK channels pharmacological treatmentof EAE animals can only partially be linked to TASK channelsexpressed on immune cells because the effects might be contami-nated througheffects ofTASKchannels expressed, e.g.onneurons(13, 23). As a note of caution, long term effects of ion channelblockers inhibiting TASK channels are not known. Therefore fur-ther work, including the detailed characterization of TASK1- andTASK3-knock out mice is clearly warranted.In summary our study introduces TASK channels on T lym-

phocytes as critical components for the maintenance of restingmembrane potential andT cell functions. Accordingly, channelmodulation results in a marked reduction of the outward cur-rent in human T cells accompanied by significantly alteredeffector functions. T cell selective pharmacological blockade inan animal model of human multiple sclerosis serves as a proofof concept concerning the (patho)physiological relevance ofthese channels. Further studies are warranted to delineate thepotential of TASK channels as a target and TASK channelblockers as potential drug candidates in T cell-mediated auto-immune diseases such as multiple sclerosis.

Acknowledgments—We thank Astrid Schmitt and Sabrina Braun-schweig for excellent technical assistance.

REFERENCES1. Schell, S. R., Nelson, D. J., Fozzard, H. A., and Fitch, F. W. (1987) J. Immu-

nol. 139, 3224–32302. Sabath, D. E., Monos, D. S., Lee, S. C., Deutsch, C., and Prystowsky, M. B.

(1986) Proc. Natl. Acad. Sci. U. S. A. 83, 4739–47433. Chandy, K. G., DeCoursey, T. E., Cahalan, M. D., McLaughlin, C., and

Gupta, S. (1984) J. Exp. Med. 160, 369–3854. Prakriya, M., and Lewis, R. S. (2003) Cell Calcium 33, 311–3215. Panyi, G., Vamosi, G., Bodnar, A., Gaspar, R., and Damjanovich, S. (2004)

Trends Immunol. 25, 565–5696. Negulescu, P. A., Shastri, N., and Cahalan, M. D. (1994) Proc. Natl. Acad.

Sci. U. S. A. 91, 2873–28777. DeCoursey, T. E., Chandy, K. G., Gupta, S., and Cahalan, M. D. (1984)

Nature 307, 465–4688. Grissmer, S., Nguyen, A. N., and Cahalan, M. D. (1993) J. Gen. Physiol.

102, 601–6309. Logsdon, N. J., Kang, J., Togo, J. A., Christian, E. P., and Aiyar, J. (1997)

J. Biol. Chem. 272, 32723–3272610. Cahalan, M. D., and Chandy, K. G. (1997) Curr. Opin. Biotechnol. 8,

749–75611. Ghanshani, S., Wulff, H., Miller, M. J., Rohm, H., Neben, A., Gutman,

G. A., Cahalan, M. D., and Chandy, K. G. (2000) J. Biol. Chem. 275,37137–37149

12. Beeton, C.,Wulff, H., Barbaria, J., Clot-Faybesse, O., Pennington, M., Ber-nard, D., Cahalan, M. D., Chandy, K. G., and Beraud, E. (2001) Proc. Natl.Acad. Sci. U. S. A. 98, 13942–13947

13. Beeton, C., Barbaria, J., Giraud, P., Devaux, J., Benoliel, A. M., Gola, M.,Sabatier, J. M., Bernard, D., Crest, M., and Beraud, E. (2001) J. Immunol.166, 936–944

14. Cahalan, M. D., Wulff, H., and Chandy, K. G. (2001) J. Clin. Immunol. 21,235–252

15. Beeton, C., and Chandy, K. G. (2005) Neuroscientist 11, 550–56216. Wulff, H., Calabresi, P. A., Allie, R., Yun, S., Pennington, M., Beeton, C.,

and Chandy, K. G. (2003) J. Clin. Investig. 111, 1703–171317. Wulff, H., Beeton, C., and Chandy, K. G. (2003) Curr. Opin. Drug Discov.

Devel. 6, 640–64718. Beeton, C., Wulff, H., Standifer, N. E., Azam, P., Mullen, K. M., Penning-

ton, M.W., Kolski-Andreaco, A., Wei, E., Grino, A., Counts, D. R., Wang,P. H., LeeHealey, C. J., Andrews, B. S., Sankaranarayanan, A., Homerick,D., Roeck, W. W., Tehranzadeh, J., Stanhope, K. L., Zimin, P., Havel, P. J.,Griffey, S., Knaus, H. G., Nepom, G. T., Gutman, G. A., Calabresi, P. A.,and Chandy, K. G. (2006) Proc. Natl. Acad. Sci. U. S. A. 103, 17414–17419

19. Kleinschnitz, C., Meuth, S. G., Kieseier, B. C., and Wiendl, H. (2006) En-docr. Metab. Immune Disord. Drug Targets 7, 35–63

20. Goldstein, S. A., Bockenhauer, D., O’Kelly, I., and Zilberberg, N. (2001)Nat. Rev. Neurosci. 2, 175–184

21. Lesage, F. (2003) Neuropharmacology 44, 1–722. Chen, W. C., and Davis, R. L. (2006) Hear. Res. 222, 89–9923. Meuth, S. G., Budde, T., Kanyshkova, T., Broicher, T., Munsch, T., and

Pape, H. C. (2003) J. Neurosci. 23, 6460–646924. Chen, X., Talley, E. M., Patel, N., Gomis, A., McIntire, W. E., Dong, B.,

Viana, F., Garrison, J. C., and Bayliss, D. A. (2006) Proc. Natl. Acad. Sci.U. S. A. 103, 3422–3427

25. Dodt, H. U., and Zieglgansberger, W. (1990) Brain Res. 537, 333–33626. Neher, E. (1992)Methods Enzymol. 207, 123–13127. Tischner, D.,Weishaupt, A., van den Brandt, J., Muller, N., Beyersdorf, N.,

Ip, C. W., Toyka, K. V., Hunig, T., Gold, R., Kerkau, T., and Reichardt,H. M. (2006) Brain 129, 2635–2647

28. Schmidt, J., Metselaar, J. M., Wauben, M. H., Toyka, K. V., Storm, G., andGold, R. (2003) Brain 126, 1895–1904

29. Hines, M. L., and Carnevale, N. T. (1997) Neural Comput. 9, 1179–120930. Schwarz, E. C., Kummerow, C., Wenning, A. S., Wagner, K., Sappok, A.,

Waggershauser, K., Griesemer, D., Strauss, B., Wolfs, M. J., Quintana, A.,and Hoth, M. (2007) Eur. J. Immunol. 37, 2723–2733

31. De Schutter, E., and Bower, J. M. (1994) J. Neurophysiol. 71, 375–40032. Teisseyre, A., and Mozrzymas, J. W. (2007) Cell Mol. Biol. Lett. 12,

220–23033. Beeton, C., Pennington, M. W., Wulff, H., Singh, S., Nugent, D., Crossley,

G., Khaytin, I., Calabresi, P. A., Chen, C. Y., Gutman, G. A., and Chandy,K. G. (2005)Mol. Pharmacol. 67, 1369–1381

34. Meuth, S. G., Kanyshkova, T., Meuth, P., Landgraf, P., Munsch, T., Lud-wig, A., Hofmann, F., Pape, H. C., and Budde, T. (2006) J. Neurophysiol. 96,1517–1529

35. Meuth, S. G., Aller, M. I., Munsch, T., Schuhmacher, T., Seidenbecher, T.,Meuth, P., Kleinschnitz, C., Pape, H. C., Wiendl, H., Wisden, W., andBudde, T. (2006)Mol. Pharmacol. 69, 1468–1476

36. Meuth, S. G., Simon, O. J., Grimm, A., Melzer, N., Herrmann, A. M.,Spitzer, P., Landgraf, P., and Wiendl, H. (2008) J. Neuroimmunol. 194,62–69

37. Rajan, S., Preisig-Muller, R., Wischmeyer, E., Nehring, R., Hanley, P. J.,Renigunta, V., Musset, B., Schlichthorl, G., Derst, C., Karschin, A., andDaut, J. (2002) J. Physiol. 545, 13–26

38. Vennekamp, J.,Wulff, H., Beeton, C., Calabresi, P. A., Grissmer, S., Hansel,W., and Chandy, K. G. (2004)Mol. Pharmacol. 65, 1364–1374

39. Teisseyre, A., and Mozrzymas, J. W. (2006) J. Physiol. Pharmacol. 57,131–147

40. Lewis, R. S., and Cahalan, M. D. (1995)Annu. Rev. Immunol. 13, 623–65341. Prakriya, M., and Lewis, R. S. (2006) J. Gen. Physiol. 128, 373–38642. Chandy, G. K., Wulff, H., Beeton, C., Pennington, M., Gutman, G. A., and

Cahalan, M. D. (2004) Trends Pharmacol. Sci. 25, 280–28943. Mathie, A. (2007) J. Physiol. 578, 377–38544. Berg, A. P., and Bayliss, D. A. (2007) J. Neurophysiol. 97, 1546–155245. Bean, B. P. (2007) Nat. Rev. Neurosci. 8, 451–46546. Kang, D., Choe, C., and Kim, D. (2004) Biochem. Biophys. Res. Commun.

323, 323–33147. Torborg, C. L., Berg, A. P., Jeffries, B. W., Bayliss, D. A., and McBain, C. J.

(2006) J. Neurosci. 26, 7362–7367

TASK on T cells

MAY 23, 2008 • VOLUME 283 • NUMBER 21 JOURNAL OF BIOLOGICAL CHEMISTRY 14569

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 12: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

48. Leonoudakis, D., Gray, A. T., Winegar, B. D., Kindler, C. H., Harada, M.,Taylor, D. M., Chavez, R. A., Forsayeth, J. R., and Yost, C. S. (1998) J. Neu-rosci. 18, 868–877

49. Brown, D. A. (2000) Curr. Biol. 10, R456–R45950. Buckler, K. J., Williams, B. A., and Honore, E. (2000) J. Physiol. 525,

135–14251. Meadows, H. J., and Randall, A. D. (2001) Neuropharmacology 40,

551–559

52. Maingret, F., Patel, A. J., Lazdunski,M., andHonore, E. (2001)EMBO J. 20,47–54

53. Musset, B., Meuth, S. G., Liu, G. X., Derst, C., Wegner, S., Pape, H. C.,Budde, T., Preisig-Muller, R., and Daut, J. (2006) J. Physiol. 572,639–657

54. Czirjak, G., and Enyedi, P. (2003)Mol. Pharmacol. 63, 646–65255. Chandy, G. K., Cahalan, M., Pennington, M., Norton, R. S., Wulff, H., and

Gutman, G. A. (2001) Toxicon 39, 1269–1276

TASK on T cells

14570 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 283 • NUMBER 21 • MAY 23, 2008

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 13: TWIK-relatedAcid-sensitiveK Channel1(TASK1)andTASK3 ... · For evaluation of TASK1/3 antibody specificity the following antibodies were used: rabbit anti-TASK1 (number P0981, Sigma),

WiendlSven G. Meuth, Stefan Bittner, Patrick Meuth, Ole J. Simon, Thomas Budde and Heinz

Influence T Lymphocyte Effector Functions Channel 1 (TASK1) and TASK3 Critically+TWIK-related Acid-sensitive K

doi: 10.1074/jbc.M800637200 originally published online March 28, 20082008, 283:14559-14570.J. Biol. Chem. 

  10.1074/jbc.M800637200Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/283/21/14559.full.html#ref-list-1

This article cites 55 references, 20 of which can be accessed free at

by guest on September 12, 2020

http://ww

w.jbc.org/

Dow

nloaded from