44
The type-2 diabetes-associated K + channel TALK-1 modulates beta-cell electrical excitability, 2 nd -phase insulin secretion, and glucose homeostasis Nicholas C. Vierra a , Prasanna K. Dadi a , Imju Jeong a , Matthew Dickerson a , David R. Powell b , and David A. Jacobson a,1 a Dept. of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232-0615, USA. b Lexicon Pharmaceuticals Inc., 8800 Technology Forest Place, The Woodlands, TX 77381, USA. 1 Correspondence should be addressed to D.A.J. Email: [email protected]. Phone: (615) 875-7655. Abbreviated title: TALK-1 channels regulate β-cell insulin secretion. Words in abstract: 151 Word in main text: 4,153 Figures: 6 figures, 1 table Supplemental Figures: 5 figures, 4 tables Page 1 of 44 Diabetes Diabetes Publish Ahead of Print, published online August 3, 2015

Page 1 of 44 Diabetes...Jul 29, 2015  · homeostasis. GSIS is coupled to Ca2+ influx, which is modulated by the orchestrated action of several ion channels. The primary glucose-sensitive

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

  • The type-2 diabetes-associated K+ channel TALK-1 modulates beta-cell electrical excitability,

    2nd

    -phase insulin secretion, and glucose homeostasis

    Nicholas C. Vierraa, Prasanna K. Dadi

    a, Imju Jeong

    a, Matthew Dickerson

    a, David R. Powell

    b,

    and David A. Jacobsona,1

    aDept. of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213

    Garland Ave., Nashville, TN 37232-0615, USA. bLexicon Pharmaceuticals Inc., 8800

    Technology Forest Place, The Woodlands, TX 77381, USA.

    1Correspondence should be addressed to D.A.J. Email: [email protected].

    Phone: (615) 875-7655.

    Abbreviated title: TALK-1 channels regulate β-cell insulin secretion.

    Words in abstract: 151

    Word in main text: 4,153

    Figures: 6 figures, 1 table

    Supplemental Figures: 5 figures, 4 tables

    Page 1 of 44 Diabetes

    Diabetes Publish Ahead of Print, published online August 3, 2015

  • Abstract

    Two-pore domain K+ (K2P) channels play an important role tuning β-cell glucose-stimulated

    insulin secretion (GSIS). The K2P channel TALK-1 is linked to type-2 diabetes risk through a

    coding sequence polymorphism (rs1535500); however, its physiological function has remained

    elusive. Here, we show that TALK-1 channels are expressed in mouse and human β-cells, where

    they serve as key regulators of electrical excitability and GSIS. We find that the rs1535500

    polymorphism, which results in an alanine to glutamate substitution in the C-terminus (Ct) of

    human TALK-1, increases channel activity. Genetic ablation of TALK-1 results in β-cell

    membrane potential (Vm) depolarization, increased islet Ca2+

    influx, and enhanced 2nd

    phase

    GSIS. Moreover, mice lacking TALK-1 channels are resistant to high-fat diet induced elevations

    in fasting glycemia. These findings reveal TALK-1 channels as important modulators of 2nd

    phase insulin secretion, and suggest a clinically relevant mechanism for rs1535500, which may

    increase type-2 diabetes risk by limiting GSIS.

    Page 2 of 44Diabetes

  • Pancreatic β-cell insulin secretion plays a central role in maintaining glucose

    homeostasis. GSIS is coupled to Ca2+

    influx, which is modulated by the orchestrated action of

    several ion channels. The primary glucose-sensitive channel of the β-cell is the ATP-sensitive K+

    channel (KATP). KATP is active under low glucose conditions, limiting insulin secretion by

    hyperpolarizing the Vm and inhibiting voltage-dependent Ca2+

    channels (VDCCs). Increased β-

    cell metabolism due to elevated glucose levels raises the intracellular ATP:ADP ratio, inhibiting

    KATP channels. The closure of KATP channels results in Vm depolarization to a plateau potential

    from which action potentials (APs) fire, allowing Ca2+

    influx through VDCCs, resulting in

    insulin secretion (1, 2). During glucose-induced KATP inhibition, the plateau potential is

    stabilized by small conductance K+ currents (3, 4), such as those mediated by two-pore domain

    K+

    (K2P) channels. For example, TWIK-related Acid Sensitive K2P (TASK-1) channels have

    been shown to polarize β-cell plateau potential suppressing Ca2+

    entry through VDCCs and

    limiting GSIS (5). However, the physiological role of the most abundant β-cell K2P channel,

    TWIK-related alkaline pH-activated K2P channel (TALK-1) remains unexplored. Because

    TALK-1 channels may regulate β-cell Ca2+

    influx and GSIS, defining their physiological role

    may illuminate therapeutic targets for treating type-2 diabetes.

    TALK-1 was originally cloned from human pancreas (6, 7). KCNK16, the gene encoding

    TALK-1 channels, is the most abundant K+

    channel transcript in mouse and human β-cells (8-

    10). Moreover, KCNK16 is the most islet-specific transcript in mice when compared to all other

    transcripts across six tissues assessed by transcriptome analysis (9). In humans, the KCNK16

    locus exhibits increased histone H3 methylation in islets compared to non-islet tissues, indicating

    that the locus is transcriptionally active in islets (11). While these observations suggest that

    Page 3 of 44 Diabetes

  • TALK-1 channels serve an important role in the islet, the physiological function(s) of TALK-1

    remains to be determined.

    The biophysical characteristics of TALK-1 have been defined in heterologous expression

    systems. These studies have revealed that TALK-1 channels produce outwardly rectifying, non-

    inactivating K+ currents which are enhanced by elevations in extracellular pH (7). Additionally,

    reactive oxygen species such as singlet oxygen have been demonstrated to increase TALK-1

    channel activity (6, 7, 12, 13). As TALK-1 currents resemble TASK-1 currents that modulate

    GSIS, TALK-1 may also play a role tuning β-cell Vm and GSIS. Nevertheless, to date there has

    been no examination of TALK-1 in primary cells, limiting our understanding of TALK-1

    channel function.

    Interestingly, genome-wide association studies have found that a non-synonymous

    polymorphism in TALK-1 (rs1535500) is associated with risk for type-2 diabetes (14-16). The

    rs1535500 polymorphism in TALK-1 results in a glutamate substitution at alanine 277 (A277E),

    in TALK-1’s cytoplasmic C-terminal tail (Ct). Given the high expression of TALK-1 in the islet,

    it has been hypothesized that polymorphisms in TALK-1 might influence hormone secretion,

    contributing to type-2 diabetes predisposition (14). The A277E polymorphism may alter K+

    currents through TALK-1, potentially perturbing β-cell Vm, Ca2+

    influx, and insulin secretion

    during the pathogenesis of type-2 diabetes. Therefore, defining the islet cell functions of TALK-

    1 channels in physiological and diabetic states is required to understand the role of

    polymorphisms in KCNK16 in the development of type-2 diabetes.

    Here, we show that TALK-1 channels are key regulators of β-cell Vm, Ca2+

    influx, and

    GSIS. We also reveal that rs1535500 increases TALK-1 channel activity, which may limit GSIS.

    These studies reveal that TALK-1 channels are important determinants of β-cell electrical

    Page 4 of 44Diabetes

  • excitability, and suggest that changes in TALK-1 activity impact GSIS and the pathogenesis of

    type-2 diabetes.

    Research Design and Methods

    Kcnk16-/-

    mouse preparation

    A Kcnk16 targeting vector was generated by inserting a 9.7 kb fragment containing exons 3-5 of

    the Kcnk16 gene (accession: NM_029006.1) into a vector containing a floxed neomycin cassette.

    The targeting vector was transfected into Protamine-Cre 129S5 ES cells. Following

    recombination, 1707 bp of the Kcnk16 gene corresponding to the 2nd base of the 119th

    codon to

    the 165th

    nt in the 3’ intron after the 5th exon were removed (Fig. S1). To identify correctly

    targeted ES cells, gDNA was isolated and digested with EcoRI, producing a 10.8 kb DNA

    fragment in WT alleles and an 8.7 kb DNA fragment in targeted alleles as assessed by Southern

    blot analysis. A correctly targeted ES cell was injected into 129S5 blastocysts, giving rise to

    germline transmission of the targeted Kcnk16 allele. Kcnk16-deficient mice were backcrossed

    with congenic C57Bl6/J mice for nine generations. All mice used were 8 – 10 weeks of age. The

    mice used for this study were handled in compliance with protocols approved by the Vanderbilt

    University Animal Care and Use Committee.

    Islet and β-cell isolation

    Islets were isolated from the pancreata of 8- to 10-week-old mice as previously described (17).

    Human islets from adult non-diabetic donors were provided by multiple isolation centers

    organized by the Integrated Islet Distribution Program (IIDP). Donor information is listed in

    Table S3. Some islets were dispersed into single cells with trituration in 0.005% trypsin and

    Page 5 of 44 Diabetes

  • cultured for 12-18 hours. Cells were maintained in RPMI 1640 with 10% FBS, 100 IU·ml−1

    penicillin, and 100 mg·ml−1

    streptomycin in a humidified incubator at 37 ◦C under an atmosphere

    of 95% air–5% CO2.

    Plasmids and transient expression

    Cells were transfected with 4 µg DNA using Lipofectamine 2000 (Life Technologies). Cells

    were co-transfected with a plasmid encoding GFP and vectors containing the coding sequence

    for human TALK-1 (accession: NM_001135106.1), or TALK-1a (accession: NM_032115.3).

    The dominant-negative TALK-1 G110E, was created by site-directed mutagenesis, and then

    cloned into a vector containing a P2A cleavage site followed by mCherry. Transfected cells were

    identified on the basis of mCherry fluorescence.

    Electrophysiological Recordings

    TALK-1 channel currents were recorded in single cells using the whole-cell patch clamp

    technique with an Axopatch 200B amplifier and pCLAMP10 software (Molecular Devices).

    Cells were washed with a Krebs–Ringer-HEPES buffer (KRB) containing (in mM): 119 NaCl, 2

    CaCl2, 4.7 KCl, 25 HEPES, 1.2 MgSO4, 1.2 KH2PO4, 11 glucose, adjusted to pH 7.35 with

    NaOH. Patch electrodes (3–5 MΩ) were loaded with intracellular solution containing (in mM)

    140 KCl, 1 MgCl2·6H2O, 10 EGTA, 10 HEPES, and 4 MgATP (pH 7.25 with KOH). Perforated

    patch recordings in intact islets were performed as previously described (18). To confirm

    recordings from human β-cells, cells were post-stained for insulin (5).

    Page 6 of 44Diabetes

  • Surface Expression Analysis

    HEK293 cells were transfected at 70% confluence with Lipofectamine 3000. After 72 hours, cell

    surface proteins were isolated using a Cell Surface Protein Isolation Kit (Pierce) according to

    manufacturer’s instructions. TALK-1 E277-FLAG and A277-FLAG isolated from the plasma

    membrane and whole cell lysates were visualized on a western blot that was probed with anti-

    FLAG M2 (Sigma) followed with HRP secondary based detection with pierce chemiluminescent

    substrate (Thermo Scientific). Immunoblot band densitometry was performed using ImageJ

    software. Surface expression for each sample was calculated as the mean band intensity of

    biotinylated protein divided by total TALK-1-FLAG.

    Measurement of Cytoplasmic Calcium

    Following overnight culture, islets were incubated for 20 minutes at 37°C in RPMI

    supplemented with 2 µM Fura-2 AM (Molecular Probes), followed by incubation in KRB with 2

    mM glucose for 20 minutes. Ca2+

    imaging was performed as previously described (5).

    Immunofluorescence Analysis

    Pancreata from 10-12 week-old mice or adult human donors (donor characteristics listed in Table

    S4) were fixed in 4% paraformaldehyde and embedded with paraffin. Rehydrated 5 µm sections

    underwent antigen retrieval using a citrate buffer according to the manufacturer’s protocol

    (Vector Labs Inc), and stained with primary antibodies against insulin (1:500, Dako), glucagon

    (1:250, Sigma) and TALK-1 (1:175, Sigma), and secondary antibodies conjugated to Cy3 and

    DyLight488 (1:300; Jackson ImmunoResearch Laboratories). Nuclei were stained using Prolong

    Page 7 of 44 Diabetes

  • Gold mountant with DAPI (Life Technologies). Sections were imaged with a Nikon Eclipse

    TE2000-U microscope and a Zeiss LSM 710 confocal microscope.

    Insulin secretion measurements

    For islet perifusion experiments, islets were allowed to recover for 24 hours following isolation

    in RPMI 1640 supplemented with 15% FBS and 11 mM glucose. GSIS was then determined by

    radioimmunoassay from perifused islets stimulated with 11 mM glucose (19). Insulin secretion

    measurements from static incubations were performed as described elsewhere (20).

    Glucose and insulin tolerance testing

    Mice were placed either on standard chow diet or high-fat diet (60 kcal% fat; Research Diets,

    Inc.) Glucose tolerance testing (GTT) and insulin tolerance testing (ITT) was performed as

    previously described (20-22).

    Statistical analyses

    Data was analyzed using pCLAMP10 or Microsoft Excel and presented as mean ± SEM.

    Statistical significance was determined using Student’s t-test.

    Results

    Pancreatic β-cells express functional TALK-1 channels.

    We first determined whether TALK-1 channels are functionally expressed in mouse β-

    cells. Using immunofluorescence staining of mouse pancreatic sections for TALK-1 and insulin,

    we found that TALK-1 was specifically expressed in the islet and co-localized with insulin-

    Page 8 of 44Diabetes

  • positive β-cellsnot α-cells (Fig. 1A). Although Kcnk16-/-

    (TALK-1 knockout [KO]) sections

    exhibited a similar staining pattern as WT sections, this was due to the recognition of the

    truncated TALK-1 protein produced by the targeted Kcnk16 allele by the TALK-1 antibody (Fig.

    S1). We next used patch clamp electrophysiology techniques to determine whether TALK-1

    currents are present in β-cells. To specifically examine K2P channels, voltage gated K+ (Kv)

    channels were blocked with TEA (10 mM), KATP channels were blocked with tolbutamide (100

    µM), and Ca2+

    was removed from the extracellular buffer to prevent activation of Ca2+

    -activated

    K+ (KCa) channels. In Kcnk16

    +/+ (WT) β-cells, an outwardly rectifying, non-inactivating K

    +

    current was observed (Fig. 1B), indicating the presence of K2P channels as previously described

    (5). K2P currents recorded from TALK-1 KO β-cells were significantly reduced ( pA/pF at +60

    mV: WT, 18.3 ± 1.2, n = 23; vs. KO, 11.7 ± 1.0, n = 25; three mice per genotype; P < 0.001)

    (Fig. 1B-C), indicating that TALK-1 forms a K+ channel in mouse β-cells and that K

    + channel

    function is not retained by truncated TALK-1 protein expressed in the KO mouse β-cells.

    We next assessed TALK-1 channel expression and currents in human β-cells. In human

    pancreas sections, we found that TALK-1 exhibited strong islet expression which co-localized

    with insulin-positive cells but not with glucagon-positive cells (Fig. 2A). Like mouse β-cells,

    human β-cells exhibit K2P currents (Fig. 2B); the cells were recorded in extracellular solution

    that blocks most other K+ channels (detailed above). To determine if the human β-cell K2P

    current includes TALK-1 currents we used a dominant-negative approach. A dominant-negative

    of TALK-1 (TALK-1 DN/P2A/mCherry) was designed by mutating the K+ selectivity filter of

    TALK-1 (TALK-1 G110E), a strategy that has been used to create dominant-negative subunits

    for other K2P channels (23). The TALK-1 DN G110E point mutation prevents channel activity

    by dimerizing with endogenous TALK-1 subunits, disrupting the channel’s K+-selectivity filter

    Page 9 of 44 Diabetes

  • and abolishing K+ flux. Additionally, the TALK-1 DN construct has a P2A cleavage sequence

    followed by a mCherry CDS downstream of TALK-1, which produces mCherry in all cells

    expressing the TALK-1 DN (24). Co-expression of TALK-1 DN/P2A/mCherry with WT TALK-

    1 in HEK293 cells resulted in near-complete suppression of TALK-1 currents, indicating that the

    TALK-1 DN inhibits TALK-1 channel activity (Fig. S2). We expressed the TALK-1 DN in

    dispersed human islet cells and recorded K2P currents from mCherry-positive cells. At the end

    of the recording, the cells were fixed and stained for insulin; only insulin-positive cells were

    analyzed. Expression of TALK-1 DN in human β-cells significantly reduced K2P currents when

    compared to cells expressing mCherry alone (pA/pF at +60 mV: mCherry, 36.7 ± 4.5, n = 10; vs.

    TALK-1 DN/P2A/mCherry, 22.1 ± 2.3, n = 11; P = 0.008; each construct was tested in β-cells

    from two donors) (Fig 2B-C). Together, these data strongly suggest that TALK-1 channels

    contribute to human β-cell K2P conductance.

    TALK-1 channel activity and surface expression is sensitive to Ct protein charge.

    The polymorphism in KCNK16 associated with type-2 diabetes risk (rs1535500) results

    in a glutamate substitution at alanine 277 in the Ct of TALK-1 (TALK-1 A277E) (Fig. 3A-B). To

    assess how this substitution influences channel function, we used site-directed mutagenesis to

    insert the A277E polymorphism in cloned human TALK-1 channels and recorded their activity

    in CHO cells. We found that TALK-1 A227E produced significantly larger whole-cell currents

    than TALK-1 A277 (Fig. 3C-D). Another non-synonymous polymorphism in strong linkage

    disequilibrium with rs1535500 is rs11756091 (14). This polymorphism is in transcript variant 2

    of KCNK16 (encoding TALK-1a), resulting in a proline substitution at histidine 301 (TALK-1a

    P301H). We recorded whole-cell currents of TALK-1a P301 and TALK-1a P301H, but found no

    Page 10 of 44Diabetes

  • significant difference in channel activity (Fig. S2). Thus, rs1535500 may reduce GSIS by

    increasing β-cell Vm polarization and reducing VDCC activity.

    To further investigate the mechanism underlying the enhanced currents produced by

    TALK-1 A277E, we performed single-channel analysis of TALK-1 A277 and TALK-1 A277E

    channels expressed in HEK293 cells (Fig. 3E). In cell-attached patches, we found that TALK-1

    A277E exhibits enhanced open probability (Po) (Po at -30 mV: TALK-1 A277, 0.09 ± 0.008; vs.

    TALK-1 A277E, 0.15 ± 0.008; P < 0.05; n = 5-6) (Fig. 3F). Unitary currents were not

    significantly different between TALK-1 A277 and TALK-1 A277E (Fig. S3). We also assessed

    how the A277E polymorphism affects channel surface localization. Surface protein biotinylation

    of HEK293 cells expressing either TALK-1 A277-FLAG or the A277E-FLAG variant

    demonstrated that TALK-1 A277E channels exhibit greater cell surface localization than TALK-

    1 A277 channels (Fig. 3G). These results indicate that TALK-1 A277E enhances channel activity

    through both elevated Po and surface localization, which would be predicted to promote β-cell Vm

    polarization and oppose GSIS.

    TALK-1 regulates ββββ-cell electrical excitability and Ca2+

    entry.

    To determine the physiological role of β-cell TALK-1 currents, we assessed how TALK-

    1 channels influence glucose-stimulated changes in β-cell Vm (Fig. 4A-B). Loss of TALK-1

    channels resulted in significant β-cell Vm depolarization over a range of glucose concentrations

    (Table 1). Furthermore, AP shape was altered in TALK-1 KO β-cells, with a tendency towards

    clustering of APs as well as reduced AP and after-hyperpolarization peak height when compared

    to WT β-cell APs (Fig. 4C-D & Table S1). TALK-1 KO β-cells also showed a reduced

    interburst interval between oscillations compared to WT β-cells (seconds: WT, 144.4 ± 21.5; vs.

    Page 11 of 44 Diabetes

  • KO, 51.4 ± 10.2; P = 0.008; n = 7) (Fig. 4G). In agreement with the reduced interburst interval,

    the plateau fraction (the ratio of time spent in the active phase to the entire period (25)) was

    significantly increased in islets lacking TALK-1 at all stimulatory glucose concentrations

    examined (Fig. 4H). Additionally, the average slope of repolarization at the termination of each

    burst was significantly less in β-cells lacking TALK-1 (mV · sec-1

    : WT, -3.95 ± 0.42; vs. KO, -

    1.25 ± 0.35; P = 0.001; n = 21) (Fig. 4E-F, I), indicating that TALK-1 channels contribute to Vm

    repolarization at the end of each oscillation of electrical activity. Together, these data show that

    TALK-1 channels modulate β-cell electrical activity.

    We next determined how changes in electrical activity caused by TALK-1 ablation

    influence glucose-stimulated islet Ca2+

    entry. When glucose concentration was increased from 2

    mM to 14 mM, control and TALK-1 KO islets exhibited oscillatory increases in [Ca2+

    ]i, (Fig.

    5A-B). We found that 2nd

    -phase Ca2+

    influx was increased (Fig. 5C), and the frequency of [Ca2+

    ]i

    oscillations in 14 mM glucose was accelerated in TALK-1 KO islets (peaks/minute: WT, 0.45 ±

    0.03, n = 136 ; vs. KO, 0.73 ± 0.05, n = 126 ; **P

  • an increased plateau fraction at 7 and 14 mM glucose, TALK-1 KO islets also secreted

    significantly more insulin than WT islets at these glucose concentrations (Fig. 5G).

    Perturbations in the frequency of islet [Ca2+

    ]i oscillation as well as total islet [Ca2+

    ]i entry

    have been demonstrated to affect insulin secretion (27, 28). To examine the contribution of

    [Ca2+

    ]i to the enhanced insulin secretion observed from TALK-1 KO islets, we “clamped”

    intracellular Ca2+

    with a depolarizing concentration of KCl (30 mM) and activated KATP currents

    with diazoxide (200 µM). When [Ca2+

    ]i was clamped in 14 mM glucose, insulin secretion from

    TALK-1-defiencient islets was comparable to WT islets (Fig 5G). These findings reveal that

    TALK-1 channel modulation of islet [Ca2+

    ]i influx plays an important role in GSIS.

    TALK-1 channels are critical for maintaining fasting glycemia.

    To assess how the increased insulin secretion caused by ablation of TALK-1 affects

    glucose homeostasis, we performed GTTs in chow-fed TALK-1 KO mice. We observed slightly

    elevated serum insulin levels in TALK-1 KO mice; however, these changes were not statistically

    significant, and we did not observe altered glucose tolerance or insulin resistance (Fig 6A-C).

    Additionally, TALK-1 KO islet morphology was similar to WT islets (Fig. S5), insulin content

    was comparable in WT and TALK-1 KO islets, and pancreatic insulin content was not different

    (Table S2). Therefore, we investigated whether the chronic metabolic stress of a high-fat diet

    (HFD) could reveal a role for TALK-1 channels in the maintenance of glucose homeostasis.

    After placing mice on a HFD, we observed protection from fasting hyperglycemia (three weeks

    on HFD; mg/dL: WT, 221.3 ± 8.4; vs. KO, 169.5 ± 5.6; P < 0.0005; n = 10) (Fig. 6D,F).

    However, there was no significant difference in insulin tolerance following exposure to a HFD

    (Fig. 6E). Furthermore, pancreatic insulin content was not significantly different between WT

    Page 13 of 44 Diabetes

  • and TALK-1 KO mice after 12 weeks on a HFD (Table S2), suggesting that the improved

    glycemia is not due to differences in β-cell proliferation. This indicates that TALK-1 channels

    play an important role in maintaining fasting glycemia under metabolically stressful conditions

    that can lead to type-2 diabetes.

    Discussion

    Physiological GSIS is dependent upon complex regulation of electrical activity by β-cell

    ion channels to control Ca2+

    influx. The K2P channel TASK-1 stabilizes the β-cell plateau

    potential, which helps tune Ca2+

    entry and GSIS (5). However, the role of TALK-1, the most

    abundant K+ channel of the β-cell, has not been determined. The results presented here

    demonstrate the role of β-cell TALK-1 channels in regulating electrical activity, Ca2+

    entry, and

    insulin secretion.

    Stimulation of islets with glucose induces [Ca2+

    ]i oscillations, which underlie pulsatile

    insulin secretion (29, 30). The frequency and duration of [Ca2+

    ]i oscillations is determined by

    alternating periods of electrical excitability (depolarization) and inactivity (hyperpolarization)

    (31). Periodic activation of a K+ current interrupts regenerative AP firing, giving rise to [Ca

    2+]i

    oscillations and pulsatile insulin secretion (32). Presently, only two K+ channels have been

    shown to contribute to this current, the KATP channel and the KCa channel of intermediate

    conductance, IK (33-35). KATP conductance fluctuates with oscillations in β-cell glucose

    metabolism and the ATP:ADP ratio, while IK is activated by elevated [Ca2+

    ]i, contributing to the

    termination of the oscillation (3, 32, 36). However, β-cell Vm and [Ca2+

    ]i oscillations persist in

    mouse islets lacking functional KATP or IK channels (33, 37). Furthermore, as IK is only briefly

    active following the reduction of [Ca2+

    ]i at the termination of the oscillation, another K+

    Page 14 of 44Diabetes

  • conductance likely helps to keep the Vm hyperpolarized between each oscillation (33). Our data

    indicate that TALK-1 provides a hyperpolarizing influence that decreases islet [Ca2+

    ]i oscillation

    frequency and plateau fraction. The greater Vm depolarization of TALK-1 KO β-cells during

    interburst phases may also explain the increased [Ca2+

    ]i oscillation frequency and elevated

    plateau fraction in TALK-1-deficient islets. Indeed, inhibition of KCa channels also results in

    interburst Vm depolarization and an increased oscillation frequency (29). Because the interburst

    Vm in TALK-1 KO β-cells is closer to the activation threshold for VDCCs, a smaller depolarizing

    stimulus would re-initiate AP firing. This is supported by recordings from preBötC neurons,

    where inhibition of TASK-1 K2P channels accelerates the frequency of AP bursting (38). [Ca2+

    ]i

    oscillation frequency and pulsatile insulin secretion are perturbed in type-2 diabetes, which is

    believed to be pathogenic (31, 39). Thus, the influence of TALK-1 channels on β-cell [Ca2+

    ]i

    oscillations could play an important role in modulating pulsatile insulin secretion.

    The ion channels that increase Ca2+

    influx during glucose-stimulated islet excitability

    also play a role in setting Ca2+

    oscillation frequency. For example, Ca2+

    -activated TRPM5

    channels increase β-cell depolarization, enhancing [Ca2+

    ]i oscillation frequency. Ablation of

    TRPM5 in mouse β-cells decreases glucose-stimulated [Ca2+

    ]i oscillations, reducing GSIS (28).

    Conversely, β-cells lacking the Ca2+

    channel β3 subunit (β3-/-

    ) show an increased [Ca2+

    ]i

    oscillation frequency and enhanced GSIS (27). Similar to observations in β3-/-

    mice, we find that

    the accelerated [Ca2+

    ]i oscillation frequency in TALK-1 KO islets is associated with an increase

    in GSIS. The accelerated [Ca2+

    ]i oscillation presumably increases GSIS; however, the increased

    plateau fraction may also amplify 2nd

    -phase insulin secretion in TALK-1 KO islets. It is well-

    known that the plateau fraction and insulin secretion increase concomitantly with elevated

    glucose concentrations (25, 40-42). Although the molecular mechanisms that modulate the

    Page 15 of 44 Diabetes

  • plateau fraction are complex, it is accepted that fluctuations in K+ conductance serve an

    important role (43). While the vast majority of information to this point has highlighted the

    importance of KATP and KCa channels in controlling the plateau fraction, our data demonstrate

    that K2P channels such as TALK-1 also serve an important role. The increase in oscillation

    frequency and plateau fraction in TALK-1 KO islets are presumably both involved in enhancing

    glucose-stimulated [Ca2+

    ]i, and 2nd

    phase insulin secretion; the exact roles of each will be

    determined in the future.

    Our data establish that the type-2 diabetes risk polymorphism rs1535500 may reduce

    GSIS by increasing TALK-1 channel activity (14, 16). The A277E substitution in the Ct of

    TALK-1 resulting from rs1535500 increases channel Po and channel surface localization. TALK-

    1 channels contribute to human β-cell K2P currents, thus, TALK-1 channels possessing the

    A277E substitution would be expected to augment β-cell K2P currents. Accordingly, A277E-

    containing TALK-1 channels would be predicted to promote Vm hyperpolarization, reducing β-

    cell excitability. Because TALK-1 channels apparently limit mouse islet basal and 2nd

    -phase

    insulin secretion, we speculate that human islets with TALK-1 A277E would exhibit diminished

    basal and 2nd

    -phase insulin secretion. Although the A277E substitution increases TALK-1

    channel activity, it is also possible that this substitution influences the mechanism(s) that

    modulate TALK-1 channels. Secretagogue-induced regulation of the Vm may differentially affect

    β-cells with TALK-1 A277E, which future studies will address. Together, these results also

    predict that gain-of-function mutations in TALK-1 channels may decrease β-cell Ca2+

    entry,

    limiting insulin secretion and leading to glucose intolerance.

    There is also the possibility that defects in TALK-1 channel function only elicit

    perturbations in glucose tolerance under the conditions of metabolic stress associated with type-2

    Page 16 of 44Diabetes

  • diabetes. Indeed, TALK-1 KO mice show reduced fasting glucose levels when placed on a HFD.

    This diet-induced phenotype reveals that TALK-1 channels play a key role in adapting to

    metabolic stress. In type-2 diabetes, defects in insulin pulsatility contribute to impaired fasting

    glycemia (44, 45), and it is thought that primary β-cell defects leading to reduced insulin

    pulsatility contribute to diabetes pathogenesis (46). We find an increase in plateau fraction and

    insulin secretion from TALK-1 KO islets at basal glucose levels (~7 mM glucose in mice).

    Interestingly, small increases in basal portal insulin have been found to suppress hepatic glucose

    production (HGP), without producing a detectable increase in peripheral insulin concentrations

    (47). We postulate that enhanced insulin delivery through the portal vein decreases basal HGP

    (44) in TALK-1 KO mice. Mice fed a HFD show increased liver insulin resistance and HGP in

    as little as three days (48). As we observe no difference in insulin tolerance between chow- or

    HFD-fed WT and TALK-1 KO mice, the elevated basal insulin secretion from TALK-1 KO

    islets is not enough to exacerbate insulin resistance. However, the increased basal insulin

    secretion from TALK-1 KO animals presumably suppresses HGP, leading to reduced fasting

    glycemia. Thus, in the context of the diabetes-linked polymorphism, TALK-1 A277E may also

    contribute to impaired fasting glycemia during the pathogenesis of type-2 diabetes by decreasing

    basal insulin secretion, which may lead to increased HGP during conditions of metabolic stress.

    Interestingly, human islets down-regulate TALK-1 expression in conditions of chronic metabolic

    stress (49), which our findings predict would increase insulin secretion. Future studies are

    required to determine how TALK-1 channels influence HGP during metabolic stress as well as in

    patients with rs1535500.

    In summary, our findings demonstrate that TALK-1 is required for normal GSIS and

    glucose homeostasis. TALK-1 channel activity hyperpolarizes the β-cell Vm, controlling Ca2+

    Page 17 of 44 Diabetes

  • entry, GSIS, and fasting glycemia. Moreover, our data show that the TALK-1 A277E

    polymorphism increases TALK-1 basal activity, predicting increased β-cell Vm

    hyperpolarization and reduced GSIS. This finding provides a molecular mechanism for

    rs1535500-linked increases in type-2 diabetes susceptibility, and suggests that inhibition of β-

    cell TALK-1 channels may be a novel therapeutic strategy to reduce hyperglycemia in type-2

    diabetes.

    Page 18 of 44Diabetes

  • Acknowledgements

    N.C.V., P.K.D., I.J., M.D., and D.A.J. performed experiments and analyzed data. N.C.V., I.J.,

    M.D., and D.A.J. designed and interpreted experiments. D.R.P. provided the Kcnk16-/-

    mouse and

    data relevant to its generation, as well as revisions on the manuscript. N.C.V. and D.A.J. wrote

    the manuscript.

    Research in the laboratory of D.A.J. was supported by NIH Grants DK081666, DK097392, and a

    Pilot and Feasibility grant through the Vanderbilt University Diabetes Research Training Center

    (P60 DK20593). N.C.V. was supported by the Vanderbilt Molecular Endocrinology Training

    Program grant 5T32DK07563. The Vanderbilt Islet Procurement and Analysis Core performed

    islet perifusion experiments, and was supported by NIH Grant DK20593. The Vanderbilt

    Hormone Assay Core performed measurements of insulin content, and was supported by NIH

    Grants DK059637 and DK020593.

    D.A.J. is the guarantor of this work and, as such, had full access to all the data in the study and

    takes responsibility for the integrity of the data and the accuracy of the data analysis.

    The authors declare no competing financial interests.

    Page 19 of 44 Diabetes

  • References

    1. Drews G, Krippeit-Drews P, Dufer M. Electrophysiology of islet cells. Advances in

    experimental medicine and biology. 2010;654:115-63. doi: 10.1007/978-90-481-3271-3_7.

    PubMed PMID: 20217497.

    2. Ashcroft FM, Rorsman P. Electrophysiology of the pancreatic β-cell. Progress in

    Biophysics and Molecular Biology. 1989;54(2):87-143. doi: http://dx.doi.org/10.1016/0079-

    6107(89)90013-8.

    3. Ren J, Sherman A, Bertram R, Goforth PB, Nunemaker CS, Waters CD, Satin LS. Slow

    oscillations of KATP conductance in mouse pancreatic islets provide support for electrical

    bursting driven by metabolic oscillations. American journal of physiology Endocrinology and

    metabolism. 2013;305(7):E805-17. doi: 10.1152/ajpendo.00046.2013. PubMed PMID:

    23921138; PubMed Central PMCID: PMC3798703.

    4. Atwater I, Ribalet B, Rojas E. Cyclic changes in potential and resistance of the beta-cell

    membrane induced by glucose in islets of Langerhans from mouse. The Journal of physiology.

    1978;278:117-39. PubMed PMID: 353252; PubMed Central PMCID: PMC1282341.

    5. Dadi PK, Vierra NC, Jacobson DA. Pancreatic beta-Cell-specific Ablation of TASK-1

    Channels Augments Glucose-stimulated Calcium Entry and Insulin Secretion, Improving

    Glucose Tolerance. Endocrinology. 2014;155(10):3757-68. doi: 10.1210/en.2013-2051. PubMed

    PMID: 24932805.

    6. Han J, Kang D, Kim D. Functional properties of four splice variants of a human

    pancreatic tandem-pore K+ channel, TALK-1. American journal of physiology Cell physiology.

    2003;285(3):C529-38. doi: 10.1152/ajpcell.00601.2002. PubMed PMID: 12724142.

    7. Girard C, Duprat F, Terrenoire C, Tinel N, Fosset M, Romey G, Lazdunski M, Lesage F.

    Genomic and functional characteristics of novel human pancreatic 2P domain K(+) channels.

    Biochemical and biophysical research communications. 2001;282(1):249-56. doi:

    10.1006/bbrc.2001.4562. PubMed PMID: 11263999.

    8. Bramswig NC, Everett LJ, Schug J, Dorrell C, Liu C, Luo Y, Streeter PR, Naji A,

    Grompe M, Kaestner KH. Epigenomic plasticity enables human pancreatic alpha to beta cell

    reprogramming. The Journal of clinical investigation. 2013;123(3):1275-84. doi:

    10.1172/JCI66514. PubMed PMID: 23434589; PubMed Central PMCID: PMC3582140.

    9. Ku GM, Kim H, Vaughn IW, Hangauer MJ, Myung Oh C, German MS, McManus MT.

    Research resource: RNA-Seq reveals unique features of the pancreatic beta-cell transcriptome.

    Molecular endocrinology. 2012;26(10):1783-92. doi: 10.1210/me.2012-1176. PubMed PMID:

    22915829; PubMed Central PMCID: PMC3458219.

    10. Eizirik DL, Sammeth M, Bouckenooghe T, Bottu G, Sisino G, Igoillo-Esteve M, Ortis F,

    Santin I, Colli ML, Barthson J, Bouwens L, Hughes L, Gregory L, Lunter G, Marselli L,

    Marchetti P, McCarthy MI, Cnop M. The human pancreatic islet transcriptome: expression of

    candidate genes for type 1 diabetes and the impact of pro-inflammatory cytokines. PLoS

    genetics. 2012;8(3):e1002552. doi: 10.1371/journal.pgen.1002552. PubMed PMID: 22412385;

    PubMed Central PMCID: PMC3297576.

    11. Stitzel ML, Sethupathy P, Pearson DS, Chines PS, Song L, Erdos MR, Welch R, Parker

    SC, Boyle AP, Scott LJ, Program NCS, Margulies EH, Boehnke M, Furey TS, Crawford GE,

    Collins FS. Global epigenomic analysis of primary human pancreatic islets provides insights into

    type 2 diabetes susceptibility loci. Cell metabolism. 2010;12(5):443-55. doi:

    Page 20 of 44Diabetes

  • 10.1016/j.cmet.2010.09.012. PubMed PMID: 21035756; PubMed Central PMCID:

    PMC3026436.

    12. Kang D, Kim D. Single-channel properties and pH sensitivity of two-pore domain K+

    channels of the TALK family. Biochemical and biophysical research communications.

    2004;315(4):836-44. doi: 10.1016/j.bbrc.2004.01.137. PubMed PMID: 14985088.

    13. Duprat F, Girard C, Jarretou G, Lazdunski M. Pancreatic two P domain K+ channels

    TALK-1 and TALK-2 are activated by nitric oxide and reactive oxygen species. The Journal of

    physiology. 2005;562(Pt 1):235-44. doi: 10.1113/jphysiol.2004.071266. PubMed PMID:

    15513946; PubMed Central PMCID: PMC1665469.

    14. Cho YS, Chen CH, Hu C, Long J, Ong RT, Sim X, Takeuchi F, Wu Y, Go MJ, Yamauchi

    T, Chang YC, Kwak SH, Ma RC, Yamamoto K, Adair LS, Aung T, Cai Q, Chang LC, Chen YT,

    Gao Y, Hu FB, Kim HL, Kim S, Kim YJ, Lee JJ, Lee NR, Li Y, Liu JJ, Lu W, Nakamura J,

    Nakashima E, Ng DP, Tay WT, Tsai FJ, Wong TY, Yokota M, Zheng W, Zhang R, Wang C, So

    WY, Ohnaka K, Ikegami H, Hara K, Cho YM, Cho NH, Chang TJ, Bao Y, Hedman AK, Morris

    AP, McCarthy MI, Consortium D, Mu TC, Takayanagi R, Park KS, Jia W, Chuang LM, Chan

    JC, Maeda S, Kadowaki T, Lee JY, Wu JY, Teo YY, Tai ES, Shu XO, Mohlke KL, Kato N, Han

    BG, Seielstad M. Meta-analysis of genome-wide association studies identifies eight new loci for

    type 2 diabetes in east Asians. Nature genetics. 2012;44(1):67-72. doi: 10.1038/ng.1019.

    PubMed PMID: 22158537; PubMed Central PMCID: PMC3582398.

    15. Replication DIG, Meta-analysis C, Asian Genetic Epidemiology Network Type 2

    Diabetes C, South Asian Type 2 Diabetes C, Mexican American Type 2 Diabetes C, Type 2

    Diabetes Genetic Exploration by Nex-generation sequencing in muylti-Ethnic Samples C,

    Mahajan A, Go MJ, Zhang W, Below JE, Gaulton KJ, Ferreira T, Horikoshi M, Johnson AD, Ng

    MC, Prokopenko I, Saleheen D, Wang X, Zeggini E, Abecasis GR, Adair LS, Almgren P, Atalay

    M, Aung T, Baldassarre D, Balkau B, Bao Y, Barnett AH, Barroso I, Basit A, Been LF, Beilby J,

    Bell GI, Benediktsson R, Bergman RN, Boehm BO, Boerwinkle E, Bonnycastle LL, Burtt N, Cai

    Q, Campbell H, Carey J, Cauchi S, Caulfield M, Chan JC, Chang LC, Chang TJ, Chang YC,

    Charpentier G, Chen CH, Chen H, Chen YT, Chia KS, Chidambaram M, Chines PS, Cho NH,

    Cho YM, Chuang LM, Collins FS, Cornelis MC, Couper DJ, Crenshaw AT, van Dam RM,

    Danesh J, Das D, de Faire U, Dedoussis G, Deloukas P, Dimas AS, Dina C, Doney AS, Donnelly

    PJ, Dorkhan M, van Duijn C, Dupuis J, Edkins S, Elliott P, Emilsson V, Erbel R, Eriksson JG,

    Escobedo J, Esko T, Eury E, Florez JC, Fontanillas P, Forouhi NG, Forsen T, Fox C, Fraser RM,

    Frayling TM, Froguel P, Frossard P, Gao Y, Gertow K, Gieger C, Gigante B, Grallert H, Grant

    GB, Grrop LC, Groves CJ, Grundberg E, Guiducci C, Hamsten A, Han BG, Hara K, Hassanali

    N, Hattersley AT, Hayward C, Hedman AK, Herder C, Hofman A, Holmen OL, Hovingh K,

    Hreidarsson AB, Hu C, Hu FB, Hui J, Humphries SE, Hunt SE, Hunter DJ, Hveem K, Hydrie ZI,

    Ikegami H, Illig T, Ingelsson E, Islam M, Isomaa B, Jackson AU, Jafar T, James A, Jia W,

    Jockel KH, Jonsson A, Jowett JB, Kadowaki T, Kang HM, Kanoni S, Kao WH, Kathiresan S,

    Kato N, Katulanda P, Keinanen-Kiukaanniemi KM, Kelly AM, Khan H, Khaw KT, Khor CC,

    Kim HL, Kim S, Kim YJ, Kinnunen L, Klopp N, Kong A, Korpi-Hyovalti E, Kowlessur S, Kraft

    P, Kravic J, Kristensen MM, Krithika S, Kumar A, Kumate J, Kuusisto J, Kwak SH, Laakso M,

    Lagou V, Lakka TA, Langenberg C, Langford C, Lawrence R, Leander K, Lee JM, Lee NR, Li

    M, Li X, Li Y, Liang J, Liju S, Lim WY, Lind L, Lindgren CM, Lindholm E, Liu CT, Liu JJ,

    Lobbens S, Long J, Loos RJ, Lu W, Luan J, Lyssenko V, Ma RC, Maeda S, Magi R, Mannisto S,

    Matthews DR, Meigs JB, Melander O, Metspalu A, Meyer J, Mirza G, Mihailov E, Moebus S,

    Mohan V, Mohlke KL, Morris AD, Muhleisen TW, Muller-Nurasyid M, Musk B, Nakamura J,

    Page 21 of 44 Diabetes

  • Nakashima E, Navarro P, Ng PK, Nica AC, Nilsson PM, Njolstad I, Nothen MM, Ohnaka K,

    Ong TH, Owen KR, Palmer CN, Pankow JS, Park KS, Parkin M, Pechlivanis S, Pedersen NL,

    Peltonen L, Perry JR, Peters A, Pinidiyapathirage JM, Platou CG, Potter S, Price JF, Qi L, Radha

    V, Rallidis L, Rasheed A, Rathman W, Rauramaa R, Raychaudhuri S, Rayner NW, Rees SD,

    Rehnberg E, Ripatti S, Robertson N, Roden M, Rossin EJ, Rudan I, Rybin D, Saaristo TE,

    Salomaa V, Saltevo J, Samuel M, Sanghera DK, Saramies J, Scott J, Scott LJ, Scott RA, Segre

    AV, Sehmi J, Sennblad B, Shah N, Shah S, Shera AS, Shu XO, Shuldiner AR, Sigurdsson G,

    Sijbrands E, Silveira A, Sim X, Sivapalaratnam S, Small KS, So WY, Stancakova A, Stefansson

    K, Steinbach G, Steinthorsdottir V, Stirrups K, Strawbridge RJ, Stringham HM, Sun Q, Suo C,

    Syvanen AC, Takayanagi R, Takeuchi F, Tay WT, Teslovich TM, Thorand B, Thorleifsson G,

    Thorsteinsdottir U, Tikkanen E, Trakalo J, Tremoli E, Trip MD, Tsai FJ, Tuomi T, Tuomilehto J,

    Uitterlinden AG, Valladares-Salgado A, Vedantam S, Veglia F, Voight BF, Wang C, Wareham

    NJ, Wennauer R, Wickremasinghe AR, Wilsgaard T, Wilson JF, Wiltshire S, Winckler W, Wong

    TY, Wood AR, Wu JY, Wu Y, Yamamoto K, Yamauchi T, Yang M, Yengo L, Yokota M,

    Young R, Zabaneh D, Zhang F, Zhang R, Zheng W, Zimmet PZ, Altshuler D, Bowden DW, Cho

    YS, Cox NJ, Cruz M, Hanis CL, Kooner J, Lee JY, Seielstad M, Teo YY, Boehnke M, Parra EJ,

    Chambers JC, Tai ES, McCarthy MI, Morris AP. Genome-wide trans-ancestry meta-analysis

    provides insight into the genetic architecture of type 2 diabetes susceptibility. Nature genetics.

    2014;46(3):234-44. doi: 10.1038/ng.2897. PubMed PMID: 24509480; PubMed Central PMCID:

    PMC3969612.

    16. Sakai K, Imamura M, Tanaka Y, Iwata M, Hirose H, Kaku K, Maegawa H, Watada H,

    Tobe K, Kashiwagi A, Kawamori R, Maeda S. Replication study for the association of 9 East

    Asian GWAS-derived loci with susceptibility to type 2 diabetes in a Japanese population. PloS

    one. 2013;8(9):e76317. doi: 10.1371/journal.pone.0076317. PubMed PMID: 24086726; PubMed

    Central PMCID: PMC3783369.

    17. Philipson LH, Rosenberg MP, Kuznetsov A, Lancaster ME, Worley JF, 3rd, Roe MW,

    Dukes ID. Delayed rectifier K+ channel overexpression in transgenic islets and beta-cells

    associated with impaired glucose responsiveness. J Biol Chem. 1994;269(45):27787-90. Epub

    1994/11/11. PubMed PMID: 7961701.

    18. Gopel SO, Kanno T, Barg S, Eliasson L, Galvanovskis J, Renstrom E, Rorsman P.

    Activation of Ca(2+)-dependent K(+) channels contributes to rhythmic firing of action potentials

    in mouse pancreatic beta cells. The Journal of general physiology. 1999;114(6):759-70. PubMed

    PMID: 10578013; PubMed Central PMCID: PMC2230648.

    19. Kang L, Dai C, Lustig ME, Bonner JS, Mayes WH, Mokshagundam S, James FD,

    Thompson CS, Lin CT, Perry CG, Anderson EJ, Neufer PD, Wasserman DH, Powers AC.

    Heterozygous SOD2 Deletion Impairs Glucose-Stimulated Insulin Secretion, but Not Insulin

    Action, in High-Fat-Fed Mice. Diabetes. 2014;63(11):3699-710. doi: 10.2337/db13-1845.

    PubMed PMID: 24947366.

    20. Dadi PK, Vierra NC, Ustione A, Piston DW, Colbran RJ, Jacobson DA. Inhibition of

    pancreatic beta-cell Ca2+/calmodulin-dependent protein kinase II reduces glucose-stimulated

    calcium influx and insulin secretion, impairing glucose tolerance. The Journal of biological

    chemistry. 2014;289(18):12435-45. doi: 10.1074/jbc.M114.562587. PubMed PMID: 24627477;

    PubMed Central PMCID: PMC4007438.

    21. Jacobson DA, Kuznetsov A, Lopez JP, Kash S, Ammala CE, Philipson LH. Kv2.1

    ablation alters glucose-induced islet electrical activity, enhancing insulin secretion. Cell

    Page 22 of 44Diabetes

  • metabolism. 2007;6(3):229-35. doi: 10.1016/j.cmet.2007.07.010. PubMed PMID: 17767909;

    PubMed Central PMCID: PMC2699758.

    22. Bao S, Jacobson DA, Wohltmann M, Bohrer A, Jin W, Philipson LH, Turk J. Glucose

    homeostasis, insulin secretion, and islet phospholipids in mice that overexpress iPLA2beta in

    pancreatic beta-cells and in iPLA2beta-null mice. American journal of physiology

    Endocrinology and metabolism. 2008;294(2):E217-29. doi: 10.1152/ajpendo.00474.2007.

    PubMed PMID: 17895289; PubMed Central PMCID: PMC2268609.

    23. Pei L, Wiser O, Slavin A, Mu D, Powers S, Jan LY, Hoey T. Oncogenic potential of

    TASK3 (Kcnk9) depends on K+ channel function. Proceedings of the National Academy of

    Sciences of the United States of America. 2003;100(13):7803-7. doi: 10.1073/pnas.1232448100.

    PubMed PMID: 12782791; PubMed Central PMCID: PMC164668.

    24. Kim JH, Lee SR, Li LH, Park HJ, Park JH, Lee KY, Kim MK, Shin BA, Choi SY. High

    cleavage efficiency of a 2A peptide derived from porcine teschovirus-1 in human cell lines,

    zebrafish and mice. PloS one. 2011;6(4):e18556. doi: 10.1371/journal.pone.0018556. PubMed

    PMID: 21602908; PubMed Central PMCID: PMC3084703.

    25. Nunemaker CS, Bertram R, Sherman A, Tsaneva-Atanasova K, Daniel CR, Satin LS.

    Glucose modulates [Ca2+]i oscillations in pancreatic islets via ionic and glycolytic mechanisms.

    Biophysical journal. 2006;91(6):2082-96. doi: 10.1529/biophysj.106.087296. PubMed PMID:

    16815907; PubMed Central PMCID: PMC1557567.

    26. Head WS, Orseth ML, Nunemaker CS, Satin LS, Piston DW, Benninger RK. Connexin-

    36 gap junctions regulate in vivo first- and second-phase insulin secretion dynamics and glucose

    tolerance in the conscious mouse. Diabetes. 2012;61(7):1700-7. doi: 10.2337/db11-1312.

    PubMed PMID: 22511206; PubMed Central PMCID: PMC3379660.

    27. Berggren PO, Yang SN, Murakami M, Efanov AM, Uhles S, Kohler M, Moede T,

    Fernstrom A, Appelskog IB, Aspinwall CA, Zaitsev SV, Larsson O, de Vargas LM, Fecher-Trost

    C, Weissgerber P, Ludwig A, Leibiger B, Juntti-Berggren L, Barker CJ, Gromada J, Freichel M,

    Leibiger IB, Flockerzi V. Removal of Ca2+ channel beta3 subunit enhances Ca2+ oscillation

    frequency and insulin exocytosis. Cell. 2004;119(2):273-84. doi: 10.1016/j.cell.2004.09.033.

    PubMed PMID: 15479643.

    28. Colsoul B, Schraenen A, Lemaire K, Quintens R, Van Lommel L, Segal A, Owsianik G,

    Talavera K, Voets T, Margolskee RF, Kokrashvili Z, Gilon P, Nilius B, Schuit FC, Vennekens

    R. Loss of high-frequency glucose-induced Ca2+ oscillations in pancreatic islets correlates with

    impaired glucose tolerance in Trpm5-/- mice. Proceedings of the National Academy of Sciences

    of the United States of America. 2010;107(11):5208-13. doi: 10.1073/pnas.0913107107.

    PubMed PMID: 20194741; PubMed Central PMCID: PMC2841940.

    29. Zhang M, Houamed K, Kupershmidt S, Roden D, Satin LS. Pharmacological properties

    and functional role of Kslow current in mouse pancreatic beta-cells: SK channels contribute to

    Kslow tail current and modulate insulin secretion. The Journal of general physiology.

    2005;126(4):353-63. doi: 10.1085/jgp.200509312. PubMed PMID: 16186562; PubMed Central

    PMCID: PMC2266621.

    30. Bergsten P, Grapengiesser E, Gylfe E, Tengholm A, Hellman B. Synchronous

    oscillations of cytoplasmic Ca2+ and insulin release in glucose-stimulated pancreatic islets. The

    Journal of biological chemistry. 1994;269(12):8749-53. PubMed PMID: 8132606.

    31. Bertram R, Sherman A, Satin LS. Electrical bursting, calcium oscillations, and

    synchronization of pancreatic islets. Advances in experimental medicine and biology.

    Page 23 of 44 Diabetes

  • 2010;654:261-79. doi: 10.1007/978-90-481-3271-3_12. PubMed PMID: 20217502; PubMed

    Central PMCID: PMC3120131.

    32. Bertram R, Sherman A, Satin LS. Metabolic and electrical oscillations: partners in

    controlling pulsatile insulin secretion. American journal of physiology Endocrinology and

    metabolism. 2007;293(4):E890-900. doi: 10.1152/ajpendo.00359.2007. PubMed PMID:

    17666486.

    33. Dufer M, Gier B, Wolpers D, Krippeit-Drews P, Ruth P, Drews G. Enhanced glucose

    tolerance by SK4 channel inhibition in pancreatic beta-cells. Diabetes. 2009;58(8):1835-43. doi:

    10.2337/db08-1324. PubMed PMID: 19401418; PubMed Central PMCID: PMC2712794.

    34. Dryselius S, Lund PE, Gylfe E, Hellman B. Variations in ATP-sensitive K+ channel

    activity provide evidence for inherent metabolic oscillations in pancreatic beta-cells.

    Biochemical and biophysical research communications. 1994;205(1):880-5. doi:

    10.1006/bbrc.1994.2746. PubMed PMID: 7999126.

    35. Larsson O, Kindmark H, Brandstrom R, Fredholm B, Berggren PO. Oscillations in

    KATP channel activity promote oscillations in cytoplasmic free Ca2+ concentration in the

    pancreatic beta cell. Proceedings of the National Academy of Sciences of the United States of

    America. 1996;93(10):5161-5. PubMed PMID: 8643546; PubMed Central PMCID: PMC39425.

    36. Kanno T, Rorsman P, Gopel SO. Glucose-dependent regulation of rhythmic action

    potential firing in pancreatic beta-cells by K(ATP)-channel modulation. The Journal of

    physiology. 2002;545(Pt 2):501-7. PubMed PMID: 12456829; PubMed Central PMCID:

    PMC2290696.

    37. Dufer M, Haspel D, Krippeit-Drews P, Aguilar-Bryan L, Bryan J, Drews G. Oscillations

    of membrane potential and cytosolic Ca(2+) concentration in SUR1(-/-) beta cells. Diabetologia.

    2004;47(3):488-98. Epub 2004/02/12. doi: 10.1007/s00125-004-1348-0. PubMed PMID:

    14872319.

    38. Koizumi H, Smerin SE, Yamanishi T, Moorjani BR, Zhang R, Smith JC. TASK channels

    contribute to the K+-dominated leak current regulating respiratory rhythm generation in vitro.

    The Journal of neuroscience : the official journal of the Society for Neuroscience.

    2010;30(12):4273-84. doi: 10.1523/JNEUROSCI.4017-09.2010. PubMed PMID: 20335463;

    PubMed Central PMCID: PMC2950010.

    39. Ravier MA, Sehlin J, Henquin JC. Disorganization of cytoplasmic Ca(2+) oscillations

    and pulsatile insulin secretion in islets from ob/ obmice. Diabetologia. 2002;45(8):1154-63. doi:

    10.1007/s00125-002-0883-9. PubMed PMID: 12189446.

    40. Beigelman PM, Ribalet B. Beta-cell electrical activity in response to high glucose

    concentration. Diabetes. 1980;29(4):263-5. PubMed PMID: 6987115.

    41. Meissner HP, Schmelz H. Membrane potential of beta-cells in pancreatic islets. Pflugers

    Archiv : European journal of physiology. 1974;351(3):195-206. PubMed PMID: 4608967.

    42. Henquin JC, Meissner HP, Schmeer W. Cyclic variations of glucose-induced electrical

    activity in pancreatic B cells. Pflugers Archiv : European journal of physiology.

    1982;393(4):322-7. PubMed PMID: 6750552.

    43. Henquin JC. D-glucose inhibits potassium efflux from pancreatic islet cells. Nature.

    1978;271(5642):271-3. PubMed PMID: 340960.

    44. Matveyenko AV, Liuwantara D, Gurlo T, Kirakossian D, Dalla Man C, Cobelli C, White

    MF, Copps KD, Volpi E, Fujita S, Butler PC. Pulsatile portal vein insulin delivery enhances

    hepatic insulin action and signaling. Diabetes. 2012;61(9):2269-79. doi: 10.2337/db11-1462.

    PubMed PMID: 22688333; PubMed Central PMCID: PMC3425431.

    Page 24 of 44Diabetes

  • 45. Hunter SJ, Atkinson AB, Ennis CN, Sheridan B, Bell PM. Association between insulin

    secretory pulse frequency and peripheral insulin action in NIDDM and normal subjects.

    Diabetes. 1996;45(5):683-6. PubMed PMID: 8621023.

    46. Hollingdal M, Juhl CB, Pincus SM, Sturis J, Veldhuis JD, Polonsky KS, Porksen N,

    Schmitz O. Failure of physiological plasma glucose excursions to entrain high-frequency

    pulsatile insulin secretion in type 2 diabetes. Diabetes. 2000;49(8):1334-40. PubMed PMID:

    10923634.

    47. Maheux P, Chen YDI, Polonsky KS, Reaven GM. Evidence that insulin can directly

    inhibit hepatic glucose production. Diabetologia. 1997;40(11):1300-6. doi: DOI

    10.1007/s001250050824. PubMed PMID: WOS:A1997YF77300009.

    48. Lee YS, Li P, Huh JY, Hwang IJ, Lu M, Kim JI, Ham M, Talukdar S, Chen A, Lu WJ,

    Bandyopadhyay GK, Schwendener R, Olefsky J, Kim JB. Inflammation is necessary for long-

    term but not short-term high-fat diet-induced insulin resistance. Diabetes. 2011;60(10):2474-83.

    doi: 10.2337/db11-0194. PubMed PMID: 21911747; PubMed Central PMCID: PMC3178297.

    49. Cnop M, Abdulkarim B, Bottu G, Cunha DA, Igoillo-Esteve M, Masini M, Turatsinze

    JV, Griebel T, Villate O, Santin I, Bugliani M, Ladriere L, Marselli L, McCarthy MI, Marchetti

    P, Sammeth M, Eizirik DL. RNA sequencing identifies dysregulation of the human pancreatic

    islet transcriptome by the saturated fatty acid palmitate. Diabetes. 2014;63(6):1978-93. doi:

    10.2337/db13-1383. PubMed PMID: 24379348.

    Page 25 of 44 Diabetes

  • Figure Legends

    Figure 1: TALK-1 is functionally expressed in mouse β-cells. (A) Immunofluorescence

    staining of TALK-1 (red) and insulin (green, upper panels) or glucagon (green, lower panels) in

    mouse pancreas sections; nuclei (blue) are shown in the merge panel. (B) Voltage-clamp

    recordings of K2P currents from isolated WT and TALK-1 KO mouse β-cells. The command

    voltage was maintained at -80 mV for 15 seconds prior to a 1-second voltage ramp from -120

    mV to +60 mV; currents are plotted between -60 and +60 mV. (C) Quantification of current

    density at -30, 0 and +60 mV in WT and TALK-1 KO mouse β-cells. Data are mean values ±

    SEM; *P

  • Quantification of current density at selected membrane potentials in CHO cells expressing

    TALK-1 A277 or TALK-1 A277E. *P < 0.05 vs. CHO; **P

  • Figure 5: [Ca2+

    ]i influx, oscillation frequency, and GSIS are increased in TALK-1 deficient

    islets. (A-B) Representative [Ca2+

    ]i recordings in islets from WT (A) and TALK-1 KO (B) mice

    stimulated with 14 mM glucose. (C) Area under the curve (AUC) quantification of 2nd

    -phase

    Ca2+

    influx in control and TALK-1 KO islets; Ca2+

    AUC was calculated in the first ~15 minutes

    of glucose-stimulated Ca2+

    -influx after regular [Ca2+

    ]i oscillations commenced (14 mM glucose).

    (D) Quantification of [Ca2+

    ]i oscillation frequency in control and TALK-1 KO islets. (E) GSIS in

    isolated WT and TALK-1 KO islets. Islets were perifused with 1 mM glucose and stimulated

    with 11 mM glucose. The insulin concentration was determined by radioimmunoassay. (F) Area

    under the curve quantification of 1st- and 2

    nd-phase insulin secretion, for periods indicated on

    graph. (G) Insulin secretion from WT and TALK-1 KO islets in static incubation; “Ca2+

    clamp”

    consisted of treatment with 14 mM glucose, 30 mM KCl, and 200 µM diazoxide. N Islet

    preparations per genotype are reported on the figure. Data are mean values ± SEM; *P < 0.05,

    **P

  • Table 1: Vm values recorded in WT and TALK-1 KO β-cells

    Recording conditions

    Interburst

    (Electrically silent)

    Vm (mV)

    Plateau

    Vm (mV)

    WT,

    2 mM glucose

    -72.7 ± 1.2

    (n = 12) n.a.

    TALK-1 KO,

    2 mM glucose

    -65.13 ± 0.66

    (n = 13) n.a.

    Statistical significance *** n.a.

    WT,

    7 mM glucose

    -64.7 ± 2.4

    (n = 9)

    -49.7 ± 1.7

    (n = 8)

    TALK-1 KO,

    7 mM glucose

    -55.7 ± 1.1

    (n = 6)

    -47.3 ± 0.8

    (n = 9)

    Statistical significance ** n.s.

    WT ,

    11 mM glucose

    -70.6 ± 2.5

    (n = 9)

    -51.2 ± 1.6

    (n = 12)

    TALK-1 KO,

    11 mM glucose

    -62.7 ± 2.1

    (n = 12)

    -44.2 ± 0.9

    (n = 13)

    Statistical significance * ***

    WT,

    14 mM glucose

    -63.1 ± 2.0

    (n = 7)

    -46.3 ± 2.2

    (n = 7)

    TALK-1 KO,

    14 mM glucose

    -54.6 ± 2.1

    (n = 7)

    -41.1 ± 0.9

    (n = 7)

    Statistical significance * *

    Table 1: Vm values recorded in WT and TALK-1 KO β-cells. The Vm of β-cells in intact WT

    and TALK-1 KO was measured under the conditions described in the table. N observations were

    made from five islet preparations per genotype. Data is presented as mean ± SEM; *P < 0.05; **P

    < 0.005; ***P < 0.0005. n.a.: not applicable; n.s.: no significant difference.

    Page 29 of 44 Diabetes

  • Figure 1: TALK-1 is functionally expressed in mouse β-cells

    -4

    0

    4

    8

    12

    16

    20

    -40 -20 0 20 40 60

    WT TALK-1 KO

    Membrane potential (mV)

    A

    B

    Cur

    rent

    den

    sity

    (pA

    /pF)

    0

    4

    8

    12

    16

    20

    -30 0 60

    WT(n = 23)TALK-1 KO(n = 25)

    ***

    *

    Membrane potential (mV)

    Cur

    rent

    den

    sity

    (pA

    /pF)

    Insulin

    Glucagon

    TALK-1

    TALK-1

    Merge

    Merge

    C

    Page 30 of 44Diabetes

  • Figure 2: Human β-cells contain functional TALK-1 channels.

    -505

    10152025303540

    -40 -20 0 20 40 60

    TALK-1 DNmCherry

    0

    10

    20

    30

    40

    -30 0 60

    mCherry(n = 10)TALK-1 DN(n = 11)

    Membrane potential (mV)

    Cur

    rent

    den

    sity

    (pA

    /pF)

    A

    B C

    *

    **

    **

    Cur

    rent

    den

    sity

    (pA

    /pF)

    Membrane potential (mV)

    Insulin

    Glucagon

    TALK-1

    TALK-1

    Merge

    Merge

    Page 31 of 44 Diabetes

  • Figure 3: TALK-1 A277E shows increased open probability and surface expression.

    -60 -30 0 30 60

    C D

    E FTALK-1 A277

    TALK-1 A277E

    20 mV

    -80 mV

    -120 mV

    Membrane potential (mV)

    *** ***

    ***

    **C

    urre

    nt d

    ensi

    ty (p

    A/p

    F)

    Cur

    rent

    den

    sity

    (pA

    /pF)

    Ope

    n pr

    obab

    ility

    Membrane potential (mV)

    ***

    -5

    0

    5

    10

    15

    20

    A B

    NC

    A277 E277N

    C

    c. CHOb. TALK-1 A277a. TALK-1 A277E a

    b

    c

    -5

    0

    5

    10

    15

    20

    25

    (n = 11) (n = 11) (n = 13)

    -120 mV-30 mV60 mV

    CHOTALK-1 A277

    TALK-1 A277E

    A277 (n = 5)A277E (n = 6)

    0

    0.05

    0.1

    0.15

    0.2

    0.25

    0 -30 -60 -80

    Extracellular

    Intracellular

    00.5

    11.5

    22.5

    33.5

    44.5

    TALK-1 A277(n = 3)

    TALK-1A277E(n = 3)

    TALK

    -1 S

    urfa

    ce E

    xpre

    ssio

    n (R

    elat

    ive

    to T

    ALK

    -1 A

    277) **

    G

    2 pA

    100 ms

    20 mV

    -80 mV

    -120 mV

    Vm

    Page 32 of 44Diabetes

  • Figure 4: TALK-1 regulates β-cell electrical activity.

    2 s-80-70-60-50-40-30-20-100

    -80-70-60-50-40-30-20-100

    60 s

    60 s500 ms

    -60-50-40-30-20-100

    -70-60-50-40-30-20-10

    -60-50-40-30-20-100

    -70-60-50-40-30-20-10

    2 s

    C E

    D F

    A C

    B D

    E

    mV

    mV

    mV mV

    mV

    mV

    WT

    TALK-1 KO

    I

    I

    Inte

    rbur

    st in

    terv

    al (s

    )

    F

    * ****

    500 ms

    I

    *

    *

    *

    n = 10 n = 10

    n = 10

    n = 5

    G H

    0

    0.2

    0.4

    0.6

    0.8

    1

    0 2 4 6 8 10 12 14

    Pla

    teau

    frac

    tion

    Glucose (mM)

    WTTALK-1 KO

    0

    30

    60

    90

    120

    150

    180

    WT TALK-1 KOn = 7 n = 7 n = 21 n = 20

    -6

    -5

    -4

    -3

    -2

    -1

    0WT TALK-1 KO

    mV

    · se

    c-1

    Page 33 of 44 Diabetes

  • Figure 5: [Ca2+]i influx, oscillation frequency, and GSIS are increased in TALK-1 deficient islets.

    0

    10

    20

    30

    40

    50

    60

    1st phase 2nd phase

    ControlTALK-1 KO

    07 mM

    glucose14 mMglucose

    14 mM glucose + 30 mM

    KCl + 200 μM Diazoxide

    60 s

    A

    **0.5

    0 F/

    F 0

    60 s

    0.50

    F/F

    0

    WT TALK-1 KO

    **

    01020104010601080110011201140

    WT TALK-1 KO

    00.30.40.50.60.70.80.9

    WT TALK-1 KO

    2nd -p

    hase

    Ca2

    + A

    UC

    Pea

    ks p

    er m

    inut

    e

    n = 136 n = 126

    n = 136 n = 126

    2nd

    Phase

    1st

    Phase

    ng in

    sulin

    /100

    IEQ

    s **

    **

    *

    *

    11 mM Glucose

    0.0

    0.5

    1.0

    1.5

    2.0

    2.5

    3.0

    0 10 20 30 40 50 60 70 80

    Insu

    lin (n

    g/10

    0 IE

    Qs/

    min

    )

    WTTALK-1 KO

    0

    0.2

    7 mMglucose

    14 mMglucose

    Ca2+ clamp

    2

    4

    6

    8

    10WTTALK-1 KO

    *

    *

    NS

    NS ng in

    sulin

    /ml/i

    slet

    /hou

    r

    (n = 4) (n = 4)(n = 4) (n = 4) (n = 7)

    Time (min)

    B C

    E F G

    D

    Page 34 of 44Diabetes

  • Figure 6: TALK-1 channels regulate fasting glycemia.

    0 20 40 60 80 100 120

    00.20.40.60.81.0

    0 10 20TALK-1 KOControl

    TALK-1 KOWT

    0

    50

    100

    150

    200

    250

    300

    0 3 6Weeks on HFD

    TALK-1 KOWT

    *

    0

    150

    200

    250

    300

    350

    400

    Blo

    od g

    luco

    se (m

    g/dL

    )

    Insu

    lin (n

    g/m

    L)

    0

    200

    250

    300

    350

    400

    450

    500

    Blo

    od g

    luco

    se (m

    g/dL

    )

    Blo

    od g

    luco

    se (m

    g/dL

    )

    ****

    E

    FD

    C

    *

    0

    20

    40

    60

    80

    100

    120

    0 20 40 60 80 100 120

    Glu

    cose

    (%) TALK-1 KOWT

    0

    20

    40

    60

    80

    100

    120

    0 20 40 60 80 100 120

    TALK-1 KOWT

    Time (min)

    Time (min)

    Glu

    cose

    (%)

    Time (min)

    0 20 40 60 80 100 120

    TALK-1 KOWT

    0

    150

    200

    250

    300

    350

    400

    450

    00.20.40.60.81.0

    0 10

    Insu

    lin (n

    g/m

    L)

    Blo

    od g

    luco

    se (m

    g/dL

    )

    Time (min)

    20

    B

    A

    0 20 40 60 80 100 120

    Page 35 of 44 Diabetes

  • The type-2 diabetes-associated K+ channel TALK-1 modulates beta-cell electrical excitability, 2nd-phase insulin secretion, and glucose homeostasis. Nicholas C. Vierra, Prasanna K. Dadi, Imju Jeong, Matthew Dickerson, David R. Powell, and David A. Jacobson

    Supplemental Figure 1

    Figure S1: Generation of TALK-1 KO mice. (A) Diagram illustrating predicted structure of Kcnk16 (TALK-1) mRNA. Exons 3-5 are removed in the targeted allele. (B) Illustration of TALK-1 protein membrane topology in WT (top) mice, and the structure of the predicted truncated TALK-1 protein in TALK-1 KO mice. Note that the antibody epitope is retained in the truncated TALK-1 protein. (C) Primer pairs used to validate expression of Kcnk16 message in WT and TALK-1 KO islets, as illustrated in panel (A). (D) Gel images of RT-PCR products obtained using primers in (C) in WT (upper image) and TALK-1 KO (lower image); note that message corresponding to exons 1-2 is expressed in TALK-1 KO islets, predicting the expression of a truncated TALK-1 protein. (E) TALK-1 transcript abundance in WT and TALK-1 KO islets, as assessed by real-time qRT-PCR; primers as in (D), expression normalized to GAPDH.

    TALK-1

    Exons 1-2

    TALK-1

    Exons 4-5

    100

    bp100

    00.20.40.60.8

    Exon 1-2 Exon 4-5

    Nor

    mal

    ized

    ex

    pres

    sion

    WT TALK-1 KO

    n.d.

    1 2 3 4 5

    TALK-1 mRNAExons 3-5 deleted in KO

    5’ 3’

    Primer pair for exons 1-2

    Primer pair for exons 4-5

    WT TALK-1 KO

    WT

    TALK-1 KO

    Extracellular

    IntracellularNC

    N

    Antibody epitope

    Extracellular

    Intracellular

    C

    Primer Sequence (5’ 3’)Productsize (bp)

    Kcnk16 Exons 1-2 (Forward)

    GACCAGCAGGCCCTGGAGCA 100

    Kcnk16 Exons 1-2 (Reverse)

    AGTCCCAGTTGCTGGGATTG 100

    Kcnk16 Exons 4-5 (Forward)

    AGCACCATTGGCTTCGGGGACTA 100

    Kcnk16 Exons 4-5 (Reverse)

    GTCCTAGGAGGATCCATATAGCT 100

    A

    B

    C

    D

    E

    Page 36 of 44Diabetes

  • Supplemental Figure 2

    Figure S2: The dominant negative TALK-1 G110E suppresses TALK-1 WT channel activity. (A) Whole-cell K2P currents in HEK293 cells expressing TALK-1 and TALK-1 G110E or a control plasmid (AmCyan) in response to a voltage ramp from -120 mV to +60 mV. Current densities are quantified in (B). Data are mean values ± SEM; ***P

  • Supplemental Figure 3

    Figure S3: TALK-1a P301H does not exhibit changes in channel activity. Whole-cell current densities at selected membrane potentials for TALK-1a WT and TALK-1a P301H expressed in CHO cells. Data are mean values ± SEM.

    -10

    -5

    0

    5

    10

    15

    20

    25

    30

    35

    40

    -120 -60 -40 -20 0 20 40 60

    Cur

    rent

    den

    sity

    (pA/

    pF)

    Membrane potential (mV)

    TALK-1a WT (n = 20)

    TALK-1a P301H (n = 21)

    Page 38 of 44Diabetes

  • Supplemental Figure 4

    Figure S4: Unitary currents are not different between TALK-1 WT and TALK-1 A277E. Single channel currents measured in on-cell patches in HEK293 cells expressing TALK-1 WT or TALK-1 A277E at indicated membrane potentials. n ≥ 5 for each point. Data are mean values ± SEM.

    -0.8

    -0.6

    -0.4

    -0.2

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    -120 -100 -80 -60 -40 -20 20

    WTA277E

    I (pA)

    Membrane potential (mV)

    Page 39 of 44 Diabetes

  • Supplemental Figure 5

    Insu

    lin/D

    AP

    ITALK-1 WT TALK-1 KO

    Figure S5: Pancreas sections from TALK-1 WT and TALK-1 KO mice. Sections were stained for insulin, as described in Research Designs and Methods.

    Page 40 of 44Diabetes

  • Table S1: Action potential characteristics in WT and TALK-1 KO β-cells

    Parameter WT (n = 7) TALK-1 KO (n = 7) P value

    Peak amplitude (mV) 37.92 ± 2.24 30.69 ± 2.24 0.04

    Antipeak amplitude (mV) 22.74 ± 1.02 18.38 ± 1.01 0.009

    Time to peak (ms) 6.28 ± 0.66 7.91 ± 1.00 0.21

    Half-width (ms) 11.77 ± 1.28 14.71 ± 1.85 0.24

    Area (mV∙ms) 395.94 ± 47.65 393.38 ± 44.53 0.96

    Instantaneous frequency (Hz) 3.92 ± 0.30 7.93 ± 2.17 0.09

    Interevent interval (ms) 323.50 ± 33.59 351.22 ± 50.45 0.66

    Event frequency (Hz) 3.27 ± 0.30 3.17 ± 0.33 0.82

    Maximum rise slope (mV/ms) 4.41 ± 0.77 3.09 ± 0.42 0.14

    Maximum decay slope (mV/ms) -5.59 ± 0.90 -3.75 ± 0.49 0.08

    Table S1: Action potential characteristics in WT and TALK-1 KO β-cells. Action potential parameters were determined over a period of 30 seconds in the second oscillation of electrical activity in islets stimulated with 14 mM glucose using the “Threshold search” event detection function in Clampfit 10 (pCLAMP 10; Molecular Devices). Data is presented as mean ± SD; Student’s t-test.

    Page 41 of 44 Diabetes

  • Table S2: Islet and pancreatic hormone content of WT and TALK-1 KO mice

    Parameter WT TALK-1 KO P value Islet insulin content (ng/IEQ)

    37.24 ± 3.31 (n = 4)

    38.87 ± 3.03 (n = 4) 0.73

    Islet glucagon content (pg/IEQ)

    912.10 ± 117.83 (n = 4)

    718.42 ± 106.95 (n = 4) 0.27

    Total pancreatic insulin (Chow diet) (ng/ml/mg tissue)

    8.73 ± 3.43 (n = 3)

    10.5 ± 4.89 (n = 3) 0.13

    Total pancreatic insulin (HFD) (ng/ml/mg tissue)

    15.56 ± 2.05 (n = 4)

    11.49 ± 1.07 (n = 4) 0.16

    Table S2: Islet and pancreatic hormone content of WT and TALK-1 KO mice. Islet hormone content was determined after perifusion experiments by RIA. Pancreatic insulin was extracted using acid ethanol, and quantified using a rodent insulin ELISA (ALPCO). Data is presented as mean ± SEM; Student’s t-test.

    Page 42 of 44Diabetes

  • Table S3: Islet donor characteristics for electrophysiology experiments using human islet cells

    Donor 1 2 Sex F M Age 32 yrs 52 yrs BMI 39.4 22.5 Ethnicity Caucasian Caucasian Type 2 Diabetes No No Table S3: Islet donor characteristics for electrophysiology experiments using human islet cells. Characteristics of human donors for islets used to examine TALK-1 currents in β-cells expressing control mCherry or the TALK-1 dominant negative construct.

    Page 43 of 44 Diabetes

  • Table S4: Human pancreas donor characteristics for immunofluorescence

    Donor 1 2 Sex M M Age 31 yrs 58 yrs Ethnicity African American CaucasianType 2 Diabetes No No

    Page 44 of 44Diabetes