Reduction in Voltage Gated K Channel_J Neurosci

Embed Size (px)

Citation preview

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    1/16

    ,

    *Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas,

    USA

    Program in Neuroscience, The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA

    Peripheral neuropathy is one of the most common compli-

    cations of diabetes. One of the most prominent features of

    diabetic neuropathy is the development of pain that typically

    involves the extremities, occurring as an exaggerated

    response to either a painful stimulus (hyperalgesia) or a

    mild and normally non-painful stimulus (allodynia) (Brown

    and Asbury 1984; Clark and Lee 1995). The precise cellular

    mechanisms of hyperalgesia and allodynia in diabetic

    neuropathic pain remain poorly understood. Increased excit-

    ability of primary sensory neurons plays a critical role in

    painful diabetic neuropathy (Hong et al. 2004; Jagodic et al.

    2007). It has been shown that voltage-gated Na+ channel

    currents are significantly increased in both small- and large-

    sized dorsal root ganglion (DRG) neurons in diabetic rats

    (Hong et al. 2004; Hong and Wiley 2006). There is also

    a significant increase in low- and high-voltage-gated

    Ca2+channel currents in DRG neurons in diabetic neuropathy

    (Hall et al. 1995; Jagodic et al. 2007). However, changes in

    other ion channels involved in the increased excitability of

    DRG neurons in diabetic neuropathic pain are not fully

    known.

    Received May 6, 2010; revised manuscript received May 26, 2010;

    accepted June 9, 2010.

    Address correspondence and reprint requests to Hui-Lin Pan, MD,

    PhD, Department of Anesthesiology and Perioperative Medicine, Unit

    110, The University of Texas MD Anderson Cancer Center, 1515 Hol-

    combe Blvd., Houston, TX 77030, USA.

    E-mail: [email protected]

    Abbreviations used: 4-AP, 4-aminopyridine; BDNF, brain-derived

    neurotrophic factor; DAP, 3,4-diaminopyridine; DRG, dorsal root

    ganglion; IB4, isolectin B4; NGF, nerve growth factor; PBS, phosphate-

    buffered saline; Kv channels, voltage-gated K+ channels; STZ,

    streptozotocin; TEA, tetraethylammonium.

    Abstract

    Abnormal hyperexcitability of primary sensory neurons plays

    an important role in neuropathic pain. Voltage-gated potas-

    sium (Kv) channels regulate neuronal excitability by affecting

    the resting membrane potential and influencing the repolari-

    zation and frequency of the action potential. In this study, we

    determined changes in Kv channels in dorsal root ganglion

    (DRG) neurons in a rat model of diabetic neuropathic pain.

    The densities of total Kv, A-type (IA) and sustained delayed

    (IK) currents were markedly reduced in medium- and large-,

    but not in small-, diameter DRG neurons in diabetic rats.

    Quantitative RT-PCR analysis revealed that the mRNA levelsof IA subunits, including Kv1.4, Kv3.4, Kv4.2, and Kv4.3, in the

    DRG were reduced 50% in diabetic rats compared with

    those in control rats. However, there were no significant dif-

    ferences in the mRNA levels of IK subunits (Kv1.1, Kv1.2,

    Kv2.1, and Kv2.2) in the DRG between the two groups.

    Incubation with brain-derived neurotrophic factor (BDNF)

    caused a large reduction in Kv currents, especially IA currents,

    in medium and large DRG neurons from control rats. Fur-

    thermore, the reductions in Kv currents and mRNA levels of IA

    subunits in diabetic rats were normalized by pre-treatment

    with anti-BDNF antibody or K252a, a TrkB tyrosine kinase

    inhibitor. In addition, the number of medium and large DRG

    neurons with BDNF immunoreactivity was greater in diabetic

    than control rats. Collectively, our findings suggest that diabe-

    tes primarily reduces Kv channel activity in medium and large

    DRG neurons. Increased BDNF activity in these neurons likely

    contributes to thereduction in Kv channel function through TrkBreceptor stimulation in painful diabetic neuropathy.

    Keywords: diabetic neuropathy, dorsal root ganglion, ion

    channels, neuropathic pain, neurotophic factors, voltage-

    gated potassium channels.

    J. Neurochem. (2010) 114, 14601475.

    JOURNAL OF NEUROCHEMISTRY | 2010 | 114 | 14601475 doi: 10.1111/j.1471-4159.2010.06863.x

    1460 Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    2010 The Authors

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    2/16

    Voltage-gated K+ (Kv) channels are important for the

    regulation of the resting membrane potential, the duration

    and frequency of the action potential, and the release of

    neurotransmitters in neurons (Kim et al. 2005; Catacuzzeno

    et al. 2008). The native Kv currents in primary sensoryneurons include two major types based on their inactivation

    kinetics and sensitivities to tetraethylammonium (TEA) and

    3,4-diaminopyridine (DAP) or 4-aminopyridine (4-AP):

    slowly inactivating delayed currents (IK) and rapidly

    inactivating transient A-type currents (IA) (Everill et al.

    1998; Liu and Simon 2003; Vydyanathan et al. 2005). In the

    isolectin B4 (IB4)-positive DRG neurons, the IA is particu-

    larly important in the control of the spike onset, the threshold

    of the action potential firing, and the firing frequency

    (Vydyanathan et al. 2005). The IK is also involved in

    determining the threshold of the action potential firing, the

    repolarization and after-hyperpolarization phase, and the

    resting potential in primary sensory neurons (Safronov et al.

    1996; Catacuzzeno et al. 2008). It has been shown that

    traumatic nerve injury reduces the mRNA levels of the

    Kv1.1, Kv1.2, Kv1.4, Kv2.2, and Kv4.2 subunits in DRG

    neurons (Kim et al. 2002) and both the IA and IK in DRG

    neurons (Everill and Kocsis 1999; Abdulla and Smith 2001;

    Yang et al. 2004). Nevertheless, little is known about the

    changes in Kv channel activity in DRG neurons in diabetic

    neuropathic pain.

    Brain-derived neurotrophic factor (BDNF) is normally

    present in some small- and medium-sized DRG neurons

    (Zhou and Rush 1996; Thompson et al. 1999). The expres-

    sion level of BDNF is increased in small-sized DRG neuronsin response to peripheral inflammation (Karchewski et al.

    2002; Obata et al. 2003a). Increased BDNF expression also

    occurs in axotomized medium and large DRG neurons

    (Tonra et al. 1998; Obata et al. 2003b) and in the DRG in

    diabetic rats (Fernyhough et al. 1995). It has been shown that

    treatment with BDNF, not nerve growth factor (NGF), can

    reduce the mRNA levels of IA subunits in DRG neurons

    (Park et al. 2003). However, it is not clear whether BDNF

    plays a role in reducing Kv channel function in DRG neurons

    in diabetic neuropathy. In this study, we (i) examined the

    changes in Kv currents in different sized DRG neurons in a

    rat model of painful diabetic neuropathy and (ii) determined

    the role of BDNF in reducing Kv channel activity in DRG

    neurons in diabetic neuropathic pain.

    Methods

    Animal model of diabetic neuropathic pain

    Male SpragueDawley rats (9 weeks old, Harlan SpragueDawley,

    Indianapolis, IN, USA) were used. All experiments were approved

    by the Animal Care and Use Committee of the University of Texas

    M. D. Anderson Cancer Center and conformed to the guidelines of

    the National Institutes of Healths Guide for the Care and Use of

    Laboratory Animals. All efforts were made to minimize both the

    suffering and number of animals used. Diabetes was induced by a

    single intraperitoneal (i.p.) injection of 60 mg/kg streptozotocin

    (STZ; Sigma, St. Louis, MO, USA) freshly dissolved in 0.9% sterile

    saline (Chen and Pan 2002). Age-matched vehicle-injected rats were

    used as the controls. Previous studies have demonstrated that after

    STZ injection, most rats display reproducible mechanical allodynia

    and hyperalgesia within 3 weeks, lasting for at least 7 weeks

    (Courteix et al. 1993; Chen and Pan 2002; Khan et al. 2002). This

    model of neuropathic pain mimics the symptoms of neuropathy in

    diabetic patients, with alterations in pain sensitivity and poor

    responses to l opioid administered systemically or intrathecally

    (Courteix et al. 1993; Malcangio and Tomlinson 1998; Zurek et al.

    2001; Chen and Pan 2002; Khan et al. 2002). Diabetes was

    confirmed in the STZ-injected rats by measurement of the blood

    glucose concentration. The glucose level in the blood from the tail

    vein was assayed using ACCU-CHEK test strips (Roche Diagnos-

    tics, Indianapolis, IN, USA). The blood glucose level was measured

    3 weeks after STZ administration, and only the rats with high blood

    glucose level (> 300 mg/dL) were used. Neuropathic pain indiabetic rats was confirmed by examination of nociceptive mechan-

    ical thresholds by using the paw pressure Analgesy-Meter (Ugo

    Basile Biological Research, Comerio, Italy) (Chen and Pan 2002,

    2006).

    Isolation of DRG neurons

    Rats were anesthetized with 23% isoflurane and then rapidly

    decapitated. The thoracic and lumbar segments of the vertebral

    column were surgically removed. The lumbar DRGs and the nerve

    roots were quickly dissected out and transferred immediately into

    DMEM (Gibco, Carlsbad, CA, USA) on ice. The DRGs were then

    dissected free of the attached connective tissues under a microscope

    and minced with fine-spring scissors. The minced ganglion

    fragments were placed in a flask containing 5 mL of DMEM inwhich trypsin (type I, 0.2 mg/mL; Sigma) and collagenase (type I,

    1 mg/mL; Sigma) had been dissolved. After incubation at 34C in a

    shaking water bath for 40 min, soybean trypsin inhibitor (type II,

    1.25 mg/mL; Sigma) was added to terminate the digestion. The cell

    suspension was subsequently plated onto a 35-mm culture dish

    containing poly-L-lysine (50 lg/mL) pre-coated coverslips and

    incubated in 5% CO2 at 37C for 1 h. The supernatant was then

    removed, and fresh DMEM was carefully added. The cells were

    then kept in the incubator for at least another hour before they were

    used for electrophysiological recordings. The final electrophysio-

    logical recordings were performed 26 h after dissociation to allow

    recovery from trypsination and mechanical disruption.

    Electrophysiological recordings

    The electrodes were pulled from GC150TF-10 glass capillaries

    (inner diameter, 1.17 mm; outer diameter, 1.5 mm; Harvard

    Apparatus, Holliston, MA, USA) using a micropipette puller and

    fire-polished. The neurons were visualized using differential

    interference contrast optics on an inverted microscope (Olympus,

    Tokyo, Japan). Images of the cells were taken with a CCD camera

    and displayed on a video monitor. The neurons were recorded in the

    whole-cell configuration at a holding potential of)90 mV using an

    EPC-10 amplifier (HEKA Instruments, Lambrecht, Germany). After

    the whole-cell configuration was established, the cell membrane

    capacitance and series resistance were electronically compensated.

    2010 The Authors

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    BDNF and K+ channels in diabetic neuropathy | 1461

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    3/16

    Leak currents were subtracted using the online P/4 protocol. All

    experiments were performed at 25C. Signals were filtered at

    1 kHz, digitized at 10 kHz, and acquired using the Pulse software

    program.

    To selectively record Kv currents and minimize the contribution

    from Ca2+and Na+ currents, the extracellular solution contained (in

    mM) 150 choline chloride, 5 KCl, 2 CaCl2, 1 MgCl2, 10 HEPES,

    1 CdCl2, and 10 D-glucose (pH 7.4 adjusted with KOH, osmolarity

    320 mOsm). Because both 4-AP and DAP can directly stimulate

    high voltage-gated Ca2+channels (Wu et al. 2009), CdCl2 was used

    to block high voltage-gated Ca2+channels. The electrode resistance

    was 23 MW when filled with the solution containing (in mM) 120

    potassium gluconate, 20 KCl, 2 MgCl2, 10 EGTA, 10 HEPES, 5

    Na2-ATP, and 1 CaCl2 (pH 7.2 adjusted with KOH, osmolarity

    300 mOsm). To further determine the Kv current subtype in the

    DRG neurons, the IA and IK currents were differentiated using

    25 mM TEA and 5 mM DAP, blockers of native IK and IA,

    respectively (Robertson and Nelson 1994; Safronov et al. 1996;

    Everill et al. 1998; Vydyanathan et al. 2005). DAP was used in thisstudy because it shows a more potent inhibition of IA than 4-AP

    (Robertson and Nelson 1994). The protocol used to measure Kv

    current activation was performed at a holding potential of)90 mV

    and consisted of 400-ms depolarization pulses from )70 to 60 mV

    in 10-mV increments at 2-s intervals (Vydyanathan et al. 2005).

    Drug application

    The drugs were dissolved in distilled water at 1000 times the final

    concentration and kept frozen in aliquots. The stock solutions were

    diluted in the appropriate external solution just before use and held

    in a series of independent syringes connected to corresponding fused

    silica columns (ID 200 lm). The distance from the column mouth to

    the cell being recorded was about 100 lm. Each drug solution was

    delivered to the recording chamber by gravity, and rapid solutionexchange (about 200 ms) was achieved by controlling the corre-

    sponding valve switch. All drugs and chemicals were purchased

    from Sigma-Aldrich except BDNF and the anti-BDNF antibody,

    which were purchased from Millipore (Temecula, CA, USA).

    Real-time RT-PCR analysis of Kv subunit expression

    Total RNA was extracted from rat lumbar DRGs at the L4-L6 level

    using the Purelink total RNA purification system (Invitrogen,

    Carlsbad, CA, USA) with on-column Dnase I digestion according to

    the manufacturers instructions. cDNA was prepared by using the

    Superscript III first-strand synthesis kit (Invitrogen).

    Quantitative PCR was performed using the iQ5 real-time PCR

    detection system with the SYBR Green PCR kit (Bio-Rad,

    Hercules, CA, USA). All samples were analyzed in duplicate using

    an annealing temperature of 60C, and each experiment was

    repeated at least once. The primer pairs used are listed in Table 1.

    To calculate the relative Kv subunit mRNA expression levels in

    each sample, standard curves were generated using a twofold

    dilution of the cDNA from the DRGs as the PCR template. The

    relative amount of Kv-subunit mRNA in each sample was first

    normalized to the level of the housekeeping gene S18 and was then

    normalized to its expression level in control rats. The PCR product

    specificity was verified by melting-curve analysis and agarose gel

    electrophoresis.

    Double immunofluorescence labeling of BDNF and Nissl in the DRG

    Four rats in each group were used for the immunofluorescence

    labeling. Rats were used 3 weeks after treatment with either STZ or

    vehicle control. Under deep anesthesia induced by sodium pento-

    barbital (60 mg/kg, i.p.), rats were perfused intracardially with

    250 mL of saline, 250 mL of 4% paraformadehyde in 0.1 M

    phosphate-buffered saline (PBS), and 150 mL of 10% sucrose in

    PBS (pH 7.4). The lumbar DRGs at the L4-L6 levels were removed

    quickly and cryoprotected in 30% sucrose in PBS for 24 h at 4C.

    To determine the distribution of BDNF in DRG neurons in both

    control and diabetic rats, immunofluorescent labeling of BDNF and

    Nissl (a neuronal marker) in the DRG sections were performed. The

    DRG tissues were cut into 30-lm-thick sections and collected free-floating in 0.1 M PBS. The tissue sections were rinsed in 0.1 M

    Tris-buffered saline, and incubated with the primary antibody (rabbit

    anti-BDNF, dilution 1 : 500; Millipore) for 2 h at 25C and then

    5 days at 4C. The specificity of this antibody has been demon-

    Table 1 List of primers used for real-time PCR

    Gene name Primer Sequence Location

    rat Kv1.1 Kv1.1-p1 5 gga gcg ccc cct acc cga gaa g 3 454475

    (NM_173095) Kv1.1-p2 5 ggt gaa tgg tgc ccg tga agt cct 3 644621

    rat Kv1.2 Kv1.2-p1 5 tcc cgg atg cct tct ggt g 3 16631683

    (NM_012970) Kv1.2-p2 5

    ggc ctg ctc ctc tcc ctc tgt 3

    18621842rat Kv1.4 Kv1.4-p1 5 ttg tga acg cgt ggt aat aaa tgt gt 3 605630

    (NM_012971) Kv1.4-p2 5 ggc ggc ctc ctg act ggt aat aat a 3 804780

    rat Kv2.1 Kv2.1-p1 5gcg act gct cag acc cct tag ctc 3 239262

    (NM_013186) Kv2.1-p2 5 tct gga atc gtg atc agc gct ttg 3 11361113

    rat Kv2.2 Kv2.2-p1 5 cgt gga gaa ggc tgg aga gtc g 3 17271748

    (NM_054000) Kv2.2-p2 5 tgg gct gga gga aga agt gtt gtt 3 19191896

    rat Kv3.4 Kv3.4-p1 5 cca cgg ggc aat gac cac acc 3 643663

    (XM_001070801) Kv3.4-p2 5 aca cag cgc acc cac cag cat tcc t 3 777753

    rat Kv4.2 Kv4.2-p1 5 gcc gca gcg cct agt cgt tac c 3 12981319

    (NM_031730) Kv4.2-p2 5 tga tag cca ttg tga ggg aaa aga gca 3 15591533

    rat Kv4.3 Kv4.3-p1 5 ctc cct aag cgg cgt cct ggt cat t 3 12531277

    (NM_031739) Kv4.3-p2 5 ctt ctg tgc cct gcg ttt atc tgc tct c 3 13611334

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    2010 The Authors

    1462 | X.-H. Cao et al.

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    4/16

    strated previously by pre-adsorption with the immunizing antigens

    (Zhou and Rush 1996; Zhou et al. 1999). The sections were rinsed

    in Tris-buffered saline and incubated with the secondary antibody

    (biotin-conjugated goat anti-rabbit IgG, dilution 1 : 200; Jackson

    ImmunoResearch Inc., West Grove, PA, USA) for 2 h at 25C.

    Then, the sections were incubated with a 1 : 100 dilution of

    streptavidin-horseradish peroxidase for 1 h at25C. Subsequently,

    the sections were incubated with fluorescein-tyramide (dilution

    1 : 100; PerkinElmer, Waltham, MA, USA) for 10 min at 25C.

    The sections were then rinsed and incubated with NeuroTrace red

    fluorescent Nissl stain (dilution 1 : 100; Molecular Probes, Eugene,

    OR, USA) for 40 min at25C. Finally, the sections were rinsed,

    Fig. 1 Reduction in the current densities of total Kv, 3,4-diamino-

    pyridine (DAP)-sensitive IA, and tetraethylammonium (TEA)-sensitive

    IK in medium dorsal root ganglion (DRG) neurons from diabetic rats.

    (a, b) Representative traces showing different types of Kv currents in

    medium DRG neurons from a control and diabetic rat. The neurons

    were held at )90 mV and depolarized from )70 to 60 mV in 10-mV

    increments (inset). (c) IV curves show the current densities of total

    Kv, IA, and IK in medium DRG neurons from rats in the control ( n = 22

    cells) and diabetic (n = 24 cells) groups. (d) Voltage-dependent

    activation kinetics (GV curves) of total Kv, IA, and IK in medium

    DRG neurons from control and diabetic rats. The V0.5 values of total

    Kv currents (control rats: 2.77 2.11 mV, n = 22; diabetic rats:

    10.57 1.71 mV, n = 25, p < 0.05) and IK (control rats: 4.85

    2.43 mV, n = 17; diabetic rats: 13.92 3.20 mV, n = 20, p < 0.05),

    but not IA (control rats: 10.18 5.59 mV, n = 19; diabetic rats: 1.35

    4.51 mV, n = 22, p > 0.05), were significantly different between the

    control and diabetic rats (t-test). There was no significant difference in

    the k value of total Kv currents (control rats: 17.75 0.75, n = 22;

    diabetic rats: 19.39 0.70, n = 25, p > 0.05), IA (control rats:

    18.10 2.21, n = 19; diabetic rats: 16.09 0.82, n = 22, p > 0.05),

    and IK (control rats: 15.05 0.59, n = 17; diabetic rats: 16.45 0.61,

    n = 20, p > 0.05) between the control and diabetic rats (t-test).

    *p < 0.05 compared with the corresponding value in the control group

    (two-way ANOVA).

    2010 The Authors

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    BDNF and K+ channels in diabetic neuropathy | 1463

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    5/16

    mounted on slides, dried, and sealed with a coverslip. The negative

    controls were processed in the same manner except the primary

    antibody was omitted. The sections were examined on a laser

    scanning confocal microscope (Carl Zeiss, Jena, Germany), and the

    areas of interest were photographed. To quantify the changes in the

    BDNF-expressing neurons from the DRG of diabetic rats, the

    number of BDNF-immunoreactive neurons and Nissl-positive

    neurons in the DRG of control and diabetic rats was counted by

    an investigator who was blinded to the experimental groups. The

    cell counting was performed using three images per tissue section,

    and three tissue sections were randomly selected from the L5 DRG

    for each rat (n = 4 rats per group).

    (a)

    (b)

    (c)

    (d)

    Fig. 2 Reduction in the current densities of total Kv, 3,4-diamino-

    pyridine (DAP)-sensitive IA, and 3,4-diaminopyridine (TEA)-sensitive

    IK in large dorsal root ganglion (DRG) neurons from diabetic rats. (a, b)

    Original traces show different types of Kv currents in large DRG

    neurons from a control and diabetic rat. Neurons were held at )90 mV

    and depolarized from )70 to 60 mV in 10-mV increments (inset). (c)

    IVcurves show differences in the current densities of total Kv, I A, and

    IK in large DRG neurons between the control (n = 19 cells) and dia-

    betic (n = 17 cells) groups. (d) Voltage-dependent activation kinetics

    of total Kv, IA, and IK in large DRG neurons from control and diabetic

    rats. The V0.5 values of total Kv currents (control: 1.38 2.21 mV,

    n = 19; diabetic: 11.98 2.66 mV, n = 17, p < 0.05) and IK (control:

    4.67 2.57 mV, n = 10; diabetic: 17.16 3.71 mV, n = 11, p < 0.05),

    but not IA (control:)4.71 3.02 mV, n = 19; diabetic: 3.33 3.97 mV,

    n = 17, p > 0.05), were significantly different between control and

    diabetic rats (t-test). There was no significant difference in the kvalue

    of total Kv currents (control: 19.66 0.95, n = 19; diabetic:

    20.47 0.75, n = 17, p > 0.05), IA (control: 16.51 1.56, n = 19;

    diabetic: 13.25 0.84, n = 17, p > 0.05), and IK (control: 16.93 1.06,

    n = 10; diabetic: 20.19 2.15, n = 11, p > 0.05) between the two

    groups (t-test). *p < 0.05 compared with the corresponding value in

    the control group (two-way ANOVA).

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    2010 The Authors

    1464 | X.-H. Cao et al.

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    6/16

    Data analysis and curve fitting

    Data are presented as mean SEM. The electrophysiological data

    were analyzed using the PulseFit software program (HEKA). The

    amplitude of the total Kv currents was measured at the peak, and the

    amplitude of the DAP-sensitive IA and TEA-sensitive IK were

    obtained by subtracting the amplitude of the DAP- and TEA-

    resistant Kv currents from that of the total Kv currents, respectively.

    The whole-cell currentvoltage (IV) curves for individual neurons

    (a)

    (b)

    (c)

    (d)

    Fig. 3 Lack of changes in the total Kv, 3,4-diaminopyridine (DAP)-

    sensitive IA, and tetraethylammonium (TEA)-sensitive IK in small dorsal

    root ganglion (DRG) neurons from diabetic rats. (a, b) Current traces

    showing different types of Kv currents in small DRG neurons from a

    control and diabetic rat. Neurons are held at )90 mV and depolarized

    from )70 to 60 mV in 10-mV increments (inset). (c) Comparison of the

    current densities of total Kv currents, IA, and IK in small DRG neurons

    between the control (n = 41 cells) and diabetic (n = 39 cells) group. (d)

    Steady-state activation (GV) curvesof total Kv, IA,andIK in small DRG

    neurons from control and diabetic rats. There was no significant dif-

    ference in the V0.5 value of total Kv currents (control: 3.80 1.98 mV,

    n = 36; diabetic: 1.01 2.02 mV, n = 39, p > 0.05), IA (control:

    10.40 4.99 mV, n = 36; diabetic: 8.58 3.81 mV, n = 37, p > 0.05),

    and IK (control: 17.29 3.06 mV, n = 23; diabetic: 13.12 2.43,

    n = 15, p > 0.05) between the control and diabetic rats ( t-test). Also,

    the k values showed no difference in total Kv currents (control:

    16.22 0.69, n = 36; diabetic: 17.01 0.71, n = 39, p > 0.05), IA

    (control: 15.29 1.55, n = 36; diabetic: 16.85 1.47, n = 37,

    p > 0.05), and IK (control: 17.53 2.31 mV, n = 23; diabetic:

    17.49 1.42, n = 15, p > 0.05) between the two groups (t-test).

    2010 The Authors

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    BDNF and K+ channels in diabetic neuropathy | 1465

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    7/16

    were generated by calculating the peak outward current at each

    testing potential and normalizing to the cell capacitance. The

    conductancevoltage (GV) curves were described with the Boltz-

    mann equation: G/Gmax = 1/[1 + exp(V0.5 ) Vm/k)], where V0.5 is

    the membrane potential at which 50% activation is observed, kis the

    slope of the function, and Vm is the membrane potential. Differences

    between the means were tested for significance using paired or

    unpaired Students t-tests, repeated-measures ANOVA followed by

    Dunnetts post hoc test, or two-way ANOVA followed by Bonferronis

    post hoc test. A p-value of < 0.05 was considered to be statistically

    significant.

    Results

    Three weeks after diabetic induction, the diabetic rats

    showed a large reduction in their paw withdrawal thresh-

    olds in response to the pressure stimulus applied to the

    hindpaw (Chen and Pan 2002; Chen et al. 2009), as

    compared with age-matched control rats (control rats:

    121.11 3.51 g; diabetic rats: 77.78 3.64 g, p < 0.05,

    t-test).

    The DRG neurons were divided into three groups

    according to their cell diameters, which were measured

    with a calibrated eyepiece reticule: small (< 30 lm),

    medium (3040 lm), and large (> 40 lm). To determine

    the whole-cell Kv currents in these three groups of DRG

    neurons, we normalized the peak outward current to the

    cell capacitance. There were no significant differences in

    the capacitance of the three groups of DRG neurons

    between the diabetic rats (small, 26.23 1.08 pF, n = 39;

    medium, 62.98 3.75 pF, n = 24; large, 114.51 6.35 pF,

    n = 17) and age-matched control rats (small, 25.77

    1.99 pF, n = 41; medium, 60.33 2.42 pF, n = 22; large,

    121.94 7.36 pF, n = 19; p < 0.05, two-way ANOVA).

    Fig. 4 Lack of differences in the total Kv,

    3,4-diaminopyridine (DAP)-sensitive IA, and

    tetraethylammonium (TEA)-sensitive IK in

    isolectin B4 (IB4)-positive and IB4-negative

    small dorsal root ganglion (DRG) neurons

    between the control and diabetic group. (a,

    b) IV curves show the similar amplitudes

    of the Kv current density in IB4-positive and

    IB4-negative neurons at different potentialsin the control (IB4-positive, n = 13 cells; IB4-

    negative, n = 5 cells) and diabetic (IB4-po-

    sitive, n = 31 cells; IB4-negative, n = 5

    cells) groups.

    Fig. 5 Changes in the mRNA levels of individual Kv subunits in the

    dorsal root ganglion (DRG) from diabetic rats. (a) Differences in the

    mRNA levelsof IA subunits(Kv1.4,Kv3.4, Kv4.2, andKv4.3) in the DRG

    between control and diabetic rats. (b) Lack of differences in the mRNA

    levels of IK subunits (Kv1.1, Kv1.2, Kv2.1, and Kv2.2) in the DRG

    between control and diabetic rats (n = 8 rats, in each group). *p < 0.05

    compared with the corresponding value in the control group (t-test).

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    2010 The Authors

    1466 | X.-H. Cao et al.

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    8/16

    (a)

    (b)

    (c)

    (d)

    (e)

    Fig. 6 Effects of brain-derived neurotrophic

    factor (BDNF) treatment on the total Kv,3,4-diaminopyridine (DAP)-sensitive IA, and

    tetraethylammonium (TEA)-sensitive IK

    currents in dorsal root ganglion (DRG)

    neurons from control rats. (a) IV curves

    show that BDNF treatment had no effect on

    the current densities of total Kv, IA, and IK in

    small DRG neurons (n = 12). (b) IVcurves

    show that BDNF treatment reduced the

    current densities of total Kv, IA, and IK in

    medium DRG neurons (n = 13). (c) IV

    curves show that reduced the current den-

    sities of total Kv, IA, and IK in large DRG

    neurons (n = 12). (d) K252a, but not K252b,

    abolished the BDNF effects on total Kv, IA,

    and IK in medium DRG neurons (n = 7 in

    each group). (e) K252a, but not K252b,

    blocked the BDNF effects on total Kv, IA,

    and IK in large DRG neurons (n = 8 in each

    group). *p < 0.05 compared with the corre-

    sponding value in the control or vehicle

    (K252b) group (two-way ANOVA). [Correction

    after online publication 27 July 2010: Figure

    6 was replaced with the correct version of

    the figure.]

    2010 The Authors

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    BDNF and K+ channels in diabetic neuropathy | 1467

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    9/16

    Reduction in Kv currents in medium and large DRG neurons

    from diabetic rats

    The total Kv current density was significantly reduced in

    diabetic rats in both medium and large DRG neurons, as

    compared with the total Kv current density in the DRGneurons from the control group (Figs 1 and 2). The peak

    current density of both the IA and IK in the medium and large

    DRG neurons was also significantly smaller in the diabetic

    than in the control group (Figs 1 and 2).

    Steady-state activation is an important property of Kv

    currents and can influence the excitability of DRG neurons.

    Thus, we determined the steady-state activation of total Kv,

    IA, and IK in medium and large DRG neurons. There was a

    significant depolarizing shift in the total Kv and IK currents

    in medium DRG neurons from diabetic rats (total Kv,

    V0.5 = 10.57 1.71 mV; IK, V0.5 = 13.92 3.20 mV), as

    compared with those from the control rats (total Kv,

    V0.5 = 2.77 2.11 mV; IK, V0.5 = 4.85 2.43 mV; t-test)

    (Fig. 1d). Also, a similar depolarizing shift was found in

    the IK in the large DRG neurons from the diabetic rats (total

    Kv, V0.5 = 11.98 2.66 mV; IK, V0.5 = 17.16 3.71 mV),

    as compared with those from the control rats (total Kv,

    V0.5 = 1.38 2.21 mV; IK, V0.5 = )4.67 2.57 mV; t-test)

    (Fig. 2d). However, there was no significant difference in the

    steady-state activation of IA in the medium and large DRG

    neurons between the diabetic and control groups (Figs 1dand 2d).

    Lack of changes in Kv currents in small DRG neurons in

    diabetic rats

    The current densities of the total Kv, IA, and IK in small DRG

    neurons from diabetic rats were not significantly different

    from the current densities in small DRG neurons from control

    rats (Fig. 3). In addition, the activation kinetics of the total,

    IA, and IK from small DRG neurons did not differ signifi-

    cantly between the control and diabetic rats (Fig. 3d).

    Because the phenotypes of the small DRG neurons are

    heterogenous, we further examined the Kv currents in IB4-

    positive and IB4-negative small DRG neurons in diabetic and

    control rats. Immediately before recording, the neurons were

    labeled with IB4Alexa Fluor 594 (3 lg/mL) in a Tyrode

    solution for 10 min and then rinsed for at least 3 min

    Fig. 7 Effects of the anti-brain-derived

    neurotrophic factor (BDNF) antibody on the

    current densities of total Kv, 3,4-diamino-

    pyridine (DAP)-sensitive IA, and tetraethy-

    lammonium (TEA)-sensitive IK currents in

    dorsal root ganglion (DRG) neurons from

    diabetic rats. (a) IV curves show that

    treatment with the anti-BDNF antibody

    (50 ng/mL) slightly increased the current

    densities of total Kv, IA, and IK in small DRG

    neurons (n = 8). (b) IV curves show that

    treatment with the anti-BDNF antibody

    profoundly increased the current densities

    of total Kv, IA, and IK in medium DRG neu-

    rons (n = 13). (c) IV curves show that

    treatment with the anti-BDNF antibody

    substantially increased the current densities

    of total Kv, IA, and IK in large DRG neurons

    (n = 9). Note that the boiled anti-BDNF

    antibody had no effects on the Kv current

    density in small, medium, or large DRG

    neurons. *p < 0.05 compared with the cor-

    responding value in the control group (two-

    way ANOVA).

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    2010 The Authors

    1468 | X.-H. Cao et al.

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    10/16

    (Vydyanathan et al. 2005). There were no significant differ-

    ences in the Kv current densities in the IB4-positive or IB4-

    negative neurons between the control and diabetic groups

    (Fig. 4).

    Changes in mRNA levels of IA and IK subunits in the DRG

    from diabetic rats

    We next measured the mRNA levels of Kv a-subunits in the

    DRG from control and diabetic rats. Because the Kv1.4,

    Kv3.4, Kv4.2, and Kv4.3 subunits contribute to the IA

    channels in DRG neurons (Stuhmer et al. 1989; Oliveret al.

    2004; Chien et al. 2007), we measured the mRNA levels of

    Kv1.4, Kv3.4, Kv4.2, and Kv4.3 in the DRG from control

    and diabetic rats. The mRNA levels of the Kv1.4, Kv3.4,

    Kv4.2, and Kv4.3 subunits were all reduced approximately

    50% in the diabetic rats, as compared with the levels in the

    control rats (Fig. 5a). Kv1.1, Kv1.2, Kv2.1, and Kv2.2 are

    important subunits of the IK channels (Murakoshi and

    Trimmer 1999; Malin and Nerbonne 2002; Beekwilder et al.

    2003). However, there were no significant differences in the

    mRNA levels of the Kv1.1, Kv1.2, Kv2.1, and Kv2.2

    subunits in the DRG between the control and diabetic rats

    (Fig. 5b).

    Role of BDNF in diabetes-induced reduction in Kv currents

    in medium and large DRG neuronsIt has been shown that BDNF expression is increased in the

    rat DRG after diabetic induction (Fernyhough et al. 1995).

    To determine whether increased BDNF contributes to the

    reduction in Kv currents in the DRG in diabetic neuropathy,

    we first determined whether BDNF treatment affects Kv

    currents in DRG neurons from control rats. Treatment of

    DRG neurons from control rats with 50 ng/mL of BDNF for

    24 h (Youssoufian and Walmsley 2007) significantly

    reduced the peak current densities of the total, IA, and IK

    in medium- and large-diameter neurons (Fig. 6ac). How-

    ever, BDNF treatment had no significant effect on the Kv

    current density in the small DRG neurons from control rats.

    TrkB is the high-affinity BDNF receptor and is primarily

    present in medium and large DRG neurons (McMahon et al.

    1994; Wetmore and Olson 1995; Karchewski et al. 1999).

    We next determined whether the effect of BDNF on Kv

    Fig. 8 Effects of K252a on the current

    densities of total Kv, 3,4-diaminopyridine

    (DAP-sensitive) IA, and tetraethylammoni-

    um (TEA)-sensitive IK currents in dorsal root

    ganglion (DRG) neurons from diabetic rats.

    (a) IV curves show that treatment with

    K252a (300 nM) slightly increased the cur-

    rent densities of total Kv, IA, and IK in small

    DRG neurons (n = 11). (b) IVcurves show

    that treatment with K252a substantially in-

    creased the current densities of total Kv, IA,

    and IK in medium DRG neurons (n = 10).

    (c) IV curves show that treatment with

    K252a profoundly increased the current

    densities of total Kv, IA, and IK in large DRG

    neurons (n = 9). *p < 0.05 compared with

    the corresponding value in the diabetic

    control group (two-way ANOVA).

    2010 The Authors

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    BDNF and K+ channels in diabetic neuropathy | 1469

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    11/16

    currents in DRG neurons was mediated by TrkB. Pre-

    treatment of DRG neurons with K252a (300 nM) (Tapley

    et al. 1992; Bhave et al. 1999), a TrkB receptor inhibitor,

    blocked the BDNF effect on the Kv current density in DRG

    neurons from control rats (Fig. 6d and e). The inactive

    analogue of K252a, K252b (300 nM), did not significantly

    alter the BDNF effect on the Kv current density in these

    neurons.

    To examine the role of BDNF in the diabetes-induced

    reduction in the Kv current density, we next tested the effect

    of the anti-BDNF antibody on the Kv currents in DRG

    neurons from diabetic rats. Incubation of DRG neurons from

    diabetic rats with the BDNF antibody (1 : 50) for 24 h

    caused a large increase in the peak current densities of the

    total Kv in all sizes of DRG neurons (Fig. 7). Also, there was

    a large increase in the IA and IKdensities in medium and large

    DRG neurons after treatment with the anti-BDNF antibody

    (Fig. 7b and c). Treatment with the boiled BDNF antibody

    had no effect on the Kv current density in DRG neurons.

    To test the hypothesis that BDNF reduces the Kv

    currents through TrkB receptor stimulation in diabetic

    neuropathy, we assessed the effect of K252a on the Kv

    current density of DRG neurons from diabetic rats.

    Incubation of DRG neurons with K252a (300 nM), for

    24 h profoundly increased the total Kv current density in

    all sizes of DRG neurons from diabetic rats (Fig. 8).

    Treatment with K252a also increased the IA and IK

    densities in all three groups of DRG neurons from diabetic

    rats (Fig. 8).

    Effects of anti-BDNF antibody and K252a on Kv currents in

    DRG neurons from control rats

    To estimate whether BDNF and its receptors (TrkB) are up-

    regulated in DRG neurons in diabetic neuropathy, we also

    examined the effects of anti-BDNF antibody and K252a on

    Kv current density in DRG neurons from control rats. In

    contrast to the evident effects of anti-BDNF antibody and

    K252a on Kv currents of DRG neurons from diabetic rats,

    treatment with BDNF antibody (1 : 50) or K252a

    (300 nM), for 24 h had little effects on the Kv current

    density in all sizes of DRG neurons obtained from control

    rats (Fig. 9).

    Fig. 9 Effects of K252a and anti-brain-

    derived neurotrophic factor (BDNF) anti-

    body on the current densities of total

    Kv, 3,4-diaminopyridine (DAP)-sensitive IA,

    and tetraethylammonium (TEA)-sensitive Ik

    currents in dorsal root ganglion (DRG)neurons from control rats. (a) IV curves

    show that treatment with K252a (300 nM,

    n = 11) and anti-BDNF antibody (50 ng/mL,

    n = 9) only had a small effect on the current

    density of IA in small DRG neurons. (b) IV

    curves show the effect of treatment with

    K252a (n = 9) and anti-BDNF antibody

    (n = 10) on the current densities of total Kv,

    IA, and Ik in medium DRG neurons. (c) IV

    curves show the lack of effect of treatment

    with K252a (n = 8) and anti-BDNF antibody

    (n = 8) on the current densities of total Kv,

    IA, and Ik in large DRG neurons. *p < 0.05

    compared with the corresponding value in

    the control group (two-way ANOVA).

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    2010 The Authors

    1470 | X.-H. Cao et al.

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    12/16

    Role of BDNF in diabetes-induced reduction in the

    expression levels of IA subunits in DRG neurons

    Because we found a large reduction in the mRNA levels of IA

    subunits in the DRG tissue of diabetic rats, we further

    determined the role of BDNF in diabetes-induced decreases

    in the expression level of Kv1.4, Kv3.4, Kv4.2, and Kv4.3

    subunits in DRG neurons using the real-time PCR technique.

    Incubation of BDNF (50 ng/mL) for 4 h in DRG neurons

    from control rats caused a large decrease in the mRNA levels

    of Kv1.4, Kv3.4, Kv4.2, and Kv4.3 subunits (Fig. 10a).

    Furthermore, in DRG neurons from diabetic rats, treatment

    with the anti-BDNF antibody (1 : 50) or K252a (300 nM)

    for 4 h reversed the decrease in the mRNA levels of Kv1.4,

    Kv3.4, Kv4.2, and Kv4.3 subunits (Fig. 10b).

    Altered distribution patterns of BDNF-immunoreactive

    DRG neurons in diabetic rats

    Additionally, we determined whether diabetic neuropathy

    affects the distribution of BDNF in different sized DRG

    neurons. In control rats, BDNF immunoreactivity was

    distributed in some small and medium DRG neurons. In

    contrast, BDNF immunoreactivity was present in most

    medium and large DRG neurons from diabetic rats

    (Fig. 11a). The total number of BDNF-immunoreactive

    neurons in the DRG was much greater in the diabetic

    (2487/4808, 51.72%) than in the control (1108/5040,21.98%; chi-squared test) rats. There were more medium

    and large neurons with BDNF immunoreactivity in the DRG

    from diabetic rats than control rats (Fig. 11b).

    Discussion

    In the present study, we found that the density of Kv currents,

    especially the IA, in medium and large DRG neurons was

    significantly reduced in a rat model of painful diabetic

    neuropathy. Quantitative PCR analysis showed that the

    mRNA levels of IA subunits, including Kv1.4, Kv3.4, Kv4.2,

    and Kv4.3, were significantly reduced in the DRG of diabetic

    rats. However, there were no significant differences in the

    mRNA levels of the IK subunits (Kv1.1, Kv1.2, Kv2.1, and

    Kv2.2) in the DRG between the diabetic and control rats.

    Furthermore, the large reduction in the Kv current density

    observed in the diabetic rats was mimicked by treatment of

    DRG neurons from control rats with BDNF. Treatment with

    either the anti-BDNF antibody or a TrkB inhibitor reversed

    the changes in the Kv current density of DRG neurons from

    diabetic rats but had little effect on Kv currents in all sizes of

    DRG neurons from control rats. In addition, the number of

    medium and large DRG neurons with BDNF immunoreac-

    tivity was markedly increased in diabetic rats. Therefore, our

    parallel biochemical and electrophysiological results provideimportant new information that diabetic neuropathy reduces

    Kv activity, particularly the IA, in medium and large DRG

    neurons. Increased BDNF activity likely contributes to the

    reduction in the Kv current density through TrkB receptor

    stimulation in diabetic neuropathy.

    Kv channels are crucial in the control of neuronal

    excitability, and their down-regulation can increase neuronal

    excitability (Pongs 1999; Vydyanathan et al. 2005; Chi and

    Nicol 2007; Chien et al. 2007; Catacuzzeno et al. 2008).

    We found here that there was a profound decrease in the

    density of total Kv, IA, and IK in medium and large DRG

    neurons from diabetic rats. In addition, the reduction in the

    IA density was greater than the reduction in the IK density in

    these DRG neurons from diabetic rats. Consistent with our

    electrophysiological data, the mRNA levels of IA subunits,

    including Kv1.4, Kv3.4, Kv4.2, and Kv4.3, were signifi-

    cantly reduced in the DRG from diabetic rats. Thus, reduced

    expression of IA subunits in diabetes could account for the

    reduced IA density seen in the medium and large DRG

    neurons from diabetic rats. Inhibition of the IA can increase

    the firing frequency and broadening of the action potential,

    leading to increased Ca2+influx and neurotransmitter release

    (Hoffman et al. 1997; Vydyanathan et al. 2005; Catacuzz-

    eno et al. 2008). For example, knockout of the Kv4.2

    Fig. 10 Reduction in the mRNA levels of IA subunits by brain-derived

    neurotrophic factor (BDNF) in dorsal root ganglion (DRG) neurons. (a)

    Effects of BDNF treatment on the mRNA levels of IA subunits (Kv1.4,

    Kv3.4, Kv4.2, and Kv4.3) in DRG neurons from control rats (n = 4

    samples in each group; t-test). (b) Effects of treatment with the anti-

    BDNF antibody (50 ng/mL) or K252a (300 nM) on the mRNA levels of

    Kv1.4, Kv3.4, Kv4.2, and Kv4.3 subunits in DRG neurons from diabetic

    rats (n = 5 samples in each group; repeated measures ANOVA).*p < 0.05 compared with the corresponding value in the control group.

    2010 The Authors

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    BDNF and K+ channels in diabetic neuropathy | 1471

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    13/16

    subunit reduces the IA and increases the excitability of DRG

    neurons, resulting in enhanced sensitivity to tactile and

    thermal stimuli (Hu et al. 2006). Furthermore, down-

    regulation of IA subunits in DRG neurons induces mechan-

    ical hypersensitivity (Chien et al. 2007). It has been shown

    that nerve ligation injury decreases the mRNA levels of the

    Kv1.1, Kv1.2, Kv1.4, Kv2.2, and Kv4.2 subunits in DRG

    neurons (Rasband et al. 2001; Kim et al. 2002). However,

    we found that although the mRNA level of Kv1.4 was

    significantly reduced in the diabetic group, the mRNA levels

    of Kv1.1 and Kv1.2 did not differ significantly between the

    control and diabetic groups. This discrepancy is likely

    caused by the difference in the peripheral nerve damage

    caused by traumatic nerve ligation and diabetic neuropathy.

    Data from our present study suggest that the reduced IA in

    medium and large DRG neurons could result from reduced

    expression of IA subunits and contributes to the abnormal

    hyperexcitability of DRG neurons in diabetic neuropathic

    pain.

    IK channels shape action potentials by keeping the single

    action potential short and elevating the firing adaptation

    (Safronov et al. 1996; Lien and Jonas 2003; Catacuzzeno

    et al. 2008). For instance, suppression of Kv1.1 by dendro-

    toxin-K or siRNA enhances the firing activity of DRG

    neurons (Chi and Nicol 2007). In addition, Kv1.1 mutant

    mice show increased pain responses (Clark and Tempel

    1998). Thus, reduction of IK channels may also contribute to

    the abnormal hyperexcitability of DRG neurons in painful

    diabetic neuropathy. We found that the IK density was

    reduced mainly in the medium and large DRG neurons from

    diabetic rats. We also observed a depolarizing shift in the IK

    in these neurons, which suggests that it is unfavorable to

    open IK channels on primary sensory neurons in diabetic

    neuropathy. However, the mRNA levels of the IK subunits,

    including Kv1.1, Kv1.2, Kv2.1, and Kv2.2, did not differ

    significantly between the control and diabetic rats. The

    mechanisms underlying the reduction in the IK in DRG

    neurons in diabetic neuropathy are not clear. The depolar-

    izing shift in the IK alone is not sufficient to explain the large

    reduction in the IK density in the medium and large DRG

    neurons from diabetic rats. Post-translational regulation,

    such as phosphorylation, may play a role in this reduction in

    the IK density in DRG neurons in diabetic rats. For example,

    the Kv channel activity reconstituted by Kv1.1 or Kv2.1 is

    controlled by phosphorylation (Boland and Jackson 1999;

    Park et al. 2006). It has been shown that diabetes increases

    the activity of protein kinase C (Ishii et al. 1998) and that

    increased protein kinase C activation can inhibit Kv1.1

    (a)

    (b)

    Fig. 11 Differences in the distribution pat-

    tern of brain-derived neurotrophic factor

    (BDNF) immunoreactive neurons in the

    dorsal root ganglion (DRG) between control

    and diabetic rats. (a) Representative con-

    focal images show a greater number of

    BDNF immunoreactive neurons in mediumand large neurons from a diabetic rat. Col-

    ocalization of BDNF and Nissl (a neuronal

    marker) is indicated in yellow when the two

    images are digitally merged. Images are

    single confocal optical sections. (b) The

    histogram shows the distinct differences in

    the distribution of BDNF immunoreactive

    neurons in different sized DRG neurons

    between control and diabetic rats. *p < 0.05

    compared with the corresponding value for

    the non-treated diabetic group (chi-squared

    test).

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    2010 The Authors

    1472 | X.-H. Cao et al.

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    14/16

    (Boland and Jackson 1999). Thus, increased protein kinase C

    activity in DRG neurons could reduce the IK density through

    phosphorylation of certain IK subunits in diabetic neuro-

    pathy.

    Previous studies of sensory neurons in diabetic neuro-pathic pain have largely focused on small DRG neurons,

    because normal nociception is thought to be mediated

    primarily by small-sized DRG neurons and unmyelinated

    afferent fibers. We have shown previously that transient

    receptor potential vanilloid 1 receptors (TRPV1)-expressing

    small sensory neurons are not involved in the development

    of allodynia in this rat model of diabetic neuropathic pain

    (Khan et al. 2002). In addition, damage to large myelinated

    afferent fibers is well known in diabetic neuropathy

    (Ochodnicka et al. 1995). We found that the diabetes-

    induced reduction in Kv currents was limited predominantly

    to medium and large DRG neurons. These findings provide

    further evidence that medium- and large-sized DRG neurons,

    which are typically associated with myelinated primary

    afferent fibers, are important in the development of diabetic

    neuropathic pain. These results indicate that the reduced Kv

    current activity in these primary sensory neurons could

    contribute to abnormal hyperexcitability of these primary

    sensory neurons and the mechanical allodynia seen in

    diabetic neuropathy.

    Another important finding of this study is that BDNF

    plays a critical role in the reduction in the Kv currents in

    DRG neurons from diabetic rats. We found that acute BDNF

    treatment reduced the mRNA levels of IA subunits in DRG

    neurons. Furthermore, BDNF treatment mainly reduced theKv current density, especially the IA current density, in

    medium and large DRG neurons from control rats. The role

    of BDNF in the reduction of Kv currents in DRG neurons

    from diabetic rats was also supported by our finding that

    treatment with the anti-BDNF antibody or a TrkB inhibitor,

    K252a, reversed the changes in the Kv currents and the

    mRNA levels of IA subunits in DRG neurons from diabetic

    rats. Our data suggest that BDNF reduces the IA current

    density by inhibiting the expression of IA subunits in DRG

    neurons in diabetic rats. Although it remains uncertain how

    BDNF suppresses the expression of IA subunits in DRG

    neurons, BDNF may inhibit the expression of IA subunits

    through the transcriptional repressor neuron restrictive

    silencing factor/repressor element-1 Silencing transcription

    factor (NRSF/REST). It has been shown that NRSF/REST is

    involved in epigenetic silencing of Kv4.3 expression by

    nerve injury in DRG neurons (Uchida et al. 2010). It is not

    clear to what degree the paradigm of BDNF application used

    in this study mimics the BDNF production in the DRG in

    diabetes. It has been suggested that the manner of BDNF

    application can have a very different effect on functional

    outcomes (Greenberg et al. 2009). Because there is no

    information about the time course of changes in BDNF

    concentrations in the DRG in diabetes, we did not compare

    whether slow and rapid increases in BDNF concentrations

    produce different effects on Kv currents in DRG neurons. It

    should be noted that K252a can block TrkA, TrkB, and

    TrkC. It has been shown that treatment with NGF (acting via

    TrkA and p75 neurotrophin receptors) can maintain Kvchannel activity after nerve ligation injury in sensory

    neurons (Everill and Kocsis 2000). However, because NGF

    does not affect the mRNA levels of IA current subunits in

    DRG neurons (Park et al. 2003), it is less likely that the

    observed effect of K252a on Kv channel activity in diabetic

    DRG neurons in this study is mediated by TrkA. We found

    that in contrast to the evident effects of anti-BDNF antibody

    and K252a on Kv currents of DRG neurons from diabetic

    rats, treatment with anti-BDNF antibody or K252a had little

    effect on the Kv current density in all sizes of DRG neurons

    from control rats. These data suggest that the functional

    activity of BDNF is increased, which reduces Kv channel

    activity through TrkB receptor stimulation in DRG neurons

    of diabetic rats.

    In addition, we found that the number of medium and large

    DRG neurons that were immunoreactive to BDNF was

    increased in diabetic rats. Our findings are consistent with the

    results from studies of rats subjected to nerve ligation injury

    (Tonra et al. 1998; Zhou et al. 1999). We found that BDNF

    treatment had little effect on the Kv currents in small DRG

    neurons in control rats. However, the Kv current density in

    small DRG neurons was slightly increased after treatment

    with the anti-BDNF antibody or a TrkB receptor inhibitor in

    the diabetic rats. Because BDNF and TrkB receptors were

    normally present in a subpopulation of small DRG neurons(McMahon et al. 1994; Zhou and Rush 1996), it is possible

    that BDNF may tonically inhibit the expression of certain Kv

    channels through TrkB receptors in these neurons in diabetic

    rats. It is not completely clear how BDNF leads to increased

    TrkB activation in DRG neurons of diabetic rats. We propose

    that BDNF released from diabetic DRG neurons activates

    neuronal TrkB through an autocrine mechanism. Therefore,

    increased BDNF activity could contribute to the large

    reduction in Kv channel function in medium and large

    DRG neurons in diabetic neuropathy through augmented

    TrkB receptor activation.

    In conclusion, this study provides novel information that

    diabetic neuropathy reduces Kv currents, particularly the IA,

    in medium and large DRG neurons. BDNF likely plays an

    important role in the reduction in the Kv channel activity of

    these neurons in painful diabetic neuropathy through TrkB

    receptor stimulation. Because reduction in Kv currents can

    enhance neuronal excitability, increased BDNF activity may

    enhance the excitability of DRG neurons by down-regulating

    the Kv channels in diabetic neuropathy. This new informa-

    tion is important for our understanding of the mechanisms

    underlying the hyperactivity of primary sensory neurons and

    the increased afferent input to the spinal dorsal horn in

    painful diabetic neuropathy.

    2010 The Authors

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    BDNF and K+ channels in diabetic neuropathy | 1473

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    15/16

    Acknowledgements

    This study was supported by the National Institutes of Health grants

    GM64830 and NS45602 and by the N.G. and Helen T. Hawkins

    Endowment to H.L.P.

    References

    Abdulla F. A. and Smith P. A. (2001) Axotomy- and autotomy-induced

    changes in Ca2+ and K+ channel currents of rat dorsal root ganglion

    neurons. J. Neurophysiol. 85, 644658.

    Beekwilder J. P., OLeary M. E., van den Broek L. P., van Kempen G. T.,

    Ypey D. L. and van den Berg R. J. (2003) Kv1.1 channels of dorsal

    root ganglion neurons are inhibited by n-butyl-p-aminobenzoate, a

    promising anesthetic for the treatment of chronic pain. J. Phar-

    macol. Exp. Ther. 304, 531538.

    Bhave S. V., Ghoda L. and Hoffman P. L. (1999) Brain-derived neuro-

    trophic factor mediates the anti-apoptotic effect of NMDA in cer-

    ebellar granule neurons: signal transduction cascades and site of

    ethanol action. J. Neurosci.19

    , 32773286.Boland L. M. and Jackson K. A. (1999) Protein kinase C inhibits Kv1.1

    potassium channel function. Am. J. Physiol. 277, C100110.

    Brown M. J. and Asbury A. K. (1984) Diabetic neuropathy. Ann. Neurol.

    15, 212.

    Catacuzzeno L., Fioretti B., Pietrobon D. and Franciolini F. (2008) The

    differential expression of low-threshold K+ currents generates

    distinct firing patterns in different subtypes of adult mouse tri-

    geminal ganglion neurones. J. Physiol. 586, 51015118.

    Chen S. R. and Pan H. L. (2002) Hypersensitivity of spinothalamic tract

    neurons associated with diabetic neuropathic pain in rats. J. Neu-

    rophysiol. 87, 27262733.

    Chen S. R. and Pan H. L. (2006) Blocking mu opioid receptors in the

    spinal cord prevents the analgesic action by subsequent systemic

    opioids. Brain Res. 1081, 119125.

    Chen S. R., Samoriski G. and Pan H. L. (2009) Antinociceptive effectsof chronic administration of uncompetitive NMDA receptor

    antagonists in a rat model of diabetic neuropathic pain. Neuro-

    pharmacology 57, 121126.

    Chi X. X. and Nicol G. D. (2007) Manipulation of the potassium channel

    Kv1.1 and its effect on neuronal excitability in rat sensory neurons.

    J. Neurophysiol. 98, 26832692.

    Chien L. Y., Cheng J. K., Chu D., Cheng C. F. and Tsaur M. L. (2007)

    Reduced expression of A-type potassium channels in primary

    sensory neurons induces mechanical hypersensitivity. J. Neurosci.

    27, 98559865.

    Clark C. M., Jr and Lee D. A. (1995) Prevention and treatment of the

    complicationsof diabetesmellitus.N. Engl. J. Med. 332, 12101217.

    Clark J. D. and Tempel B. L. (1998) Hyperalgesia in mice lacking the

    Kv1.1 potassium channel gene. Neurosci. Lett. 251, 121124.

    Courteix C., Eschalier A. and Lavarenne J. (1993) Streptozocin-induceddiabetic rats: behavioural evidence for a model of chronic pain.

    Pain 53, 8188.

    Everill B. and Kocsis J. D. (1999) Reduction in potassium currents in

    identified cutaneous afferent dorsal root ganglion neurons after

    axotomy. J. Neurophysiol. 82, 700708.

    Everill B. and Kocsis J. D. (2000) Nerve growth factor maintains

    potassium conductance after nerve injury in adult cutaneous

    afferent dorsal root ganglion neurons. Neuroscience 100, 417

    422.

    Everill B., Rizzo M. A. and Kocsis J. D. (1998) Morphologically

    identified cutaneous afferent DRG neurons express three different

    potassium currents in varying proportions. J. Neurophysiol. 79,

    18141824.

    Fernyhough P., Diemel L. T., Brewster W. J. and Tomlinson D. R. (1995)

    Altered neurotrophin mRNA levels in peripheral nerve and skeletal

    muscle of experimentally diabetic rats. J. Neurochem. 64, 1231

    1237.

    Greenberg M. E., Xu B., Lu B. and Hempstead B. L. (2009) New

    insights in the biology of BDNF synthesis and release: implicationsin CNS function. J. Neurosci. 29, 1276412767.

    Hall K. E., Browning M. D., Dudek E. M. and Macdonald R. L. (1995)

    Enhancement of high threshold calcium currents in rat primary

    afferent neurons by constitutively active protein kinase C.

    J. Neurosci. 15, 60696076.

    Hoffman D. A., Magee J. C., Colbert C. M. and Johnston D. (1997)

    K+ channel regulation of signal propagation in dendrites of

    hippocampal pyramidal neurons. Nature 387, 869875.

    Hong S. and Wiley J. W. (2006) Altered expression and function of

    sodium channels in large DRG neurons and myelinated A-fibers in

    early diabetic neuropathy in the rat. Biochem. Biophys. Res.

    Commun. 339, 652660.

    Hong S., Morrow T. J., Paulson P. E., Isom L. L. and Wiley J. W. (2004)

    Early painful diabetic neuropathy is associated with differential

    changes in tetrodotoxin-sensitive and -resistant sodium channels indorsal root ganglion neurons in the rat. J. Biol. Chem. 279, 29341

    29350.

    Hu H. J., Carrasquillo Y., Karim F., Jung W. E., Nerbonne J. M.,

    Schwarz T. L. and Gereau R. W. t. (2006) The kv4.2 potas-

    sium channel subunit is required for pain plasticity. Neuron 50,

    89100.

    Ishii H., Koya D. and King G. L. (1998) Protein kinase C activation and

    its role in the development of vascular complications in diabetes

    mellitus. J. Mol. Med. 76, 2131.

    Jagodic M. M., Pathirathna S., Nelson M. T., Mancuso S., Joksovic

    P. M., Rosenberg E. R., Bayliss D. A., Jevtovic-Todorovic V. and

    Todorovic S. M. (2007) Cell-specific alterations of T-type calcium

    current in painful diabetic neuropathy enhance excitability of

    sensory neurons. J. Neurosci. 27, 33053316.

    Karchewski L. A., Kim F. A., Johnston J., McKnight R. M. and VergeV. M. (1999) Anatomical evidence supporting the potential for

    modulation by multiple neurotrophins in the majority of adult

    lumbar sensory neurons. J. Comp. Neurol. 413, 327341.

    Karchewski L. A., Gratto K. A., Wetmore C. and Verge V. M. (2002)

    Dynamic patterns of BDNF expression in injured sensory neurons:

    differential modulation by NGF and NT-3. Eur. J. Neurosci. 16,

    14491462.

    Khan G. M., Chen S. R. and Pan H. L. (2002) Role of primary afferent

    nerves in allodynia caused by diabetic neuropathy in rats. Neuro-

    science 114, 291299.

    Kim D. S., Choi J. O., Rim H. D. and Cho H. J. (2002) Downregulation

    of voltage-gated potassium channel alpha gene expression in dorsal

    root ganglia following chronic constriction injury of the rat sciatic

    nerve. Brain Res. Mol. Brain Res. 105, 146152.

    Kim J., Wei D. S. and Hoffman D. A. (2005) Kv4 potassium channelsubunits control action potential repolarization and frequency-

    dependent broadening in rat hippocampal CA1 pyramidal neuro-

    nes. J. Physiol. 569, 4157.

    Lien C. C. and Jonas P. (2003) Kv3 potassium conductance is necessary

    and kinetically optimized for high-frequency action potential

    generation in hippocampal interneurons. J. Neurosci. 23, 2058

    2068.

    Liu L. and Simon S. A. (2003) Modulation of IA currents by capsaicin in

    rat trigeminal ganglion neurons. J. Neurophysiol. 89, 13871401.

    Malcangio M. and Tomlinson D. R. (1998) A pharmacologic analysis of

    mechanical hyperalgesia in streptozotocin/diabetic rats. Pain 76,

    151157.

    Journal Compilation 2010 International Society for Neurochemistry, J. Neurochem. (2010) 114, 14601475

    2010 The Authors

    1474 | X.-H. Cao et al.

  • 8/2/2019 Reduction in Voltage Gated K Channel_J Neurosci

    16/16

    Malin S. A. and Nerbonne J. M. (2002) Delayed rectifier K+ currents,

    IK, are encoded by Kv2 alpha-subunits and regulate tonic firing in

    mammalian sympathetic neurons. J. Neurosci. 22, 1009410105.

    McMahon S. B., Armanini M. P., Ling L. H. and Phillips H. S. (1994)

    Expression and coexpression of Trk receptors in subpopulations of

    adult primary sensory neurons projecting to identified peripheraltargets. Neuron 12, 11611171.

    Murakoshi H. and Trimmer J. S. (1999) Identification of the Kv2.1 K+

    channel as a major component of the delayed rectifier K+ current in

    rat hippocampal neurons. J. Neurosci. 19, 17281735.

    Obata K., Yamanaka H., Dai Y., Tachibana T., Fukuoka T., Tokunaga A.,

    Yoshikawa H. and Noguchi K. (2003a) Differential activation of

    extracellular signal-regulated protein kinase in primary afferent

    neurons regulates brain-derived neurotrophic factor expression

    after peripheral inflammation and nerve injury. J. Neurosci. 23,

    41174126.

    Obata K., Yamanaka H., Fukuoka T., Yi D., Tokunaga A., Hashimoto N.,

    Yoshikawa H. and Noguchi K. (2003b) Contribution of injured and

    uninjured dorsal root ganglion neurons to pain behavior and the

    changes in gene expression following chronic constriction injury of

    the sciatic nerve in rats. Pain 101, 6577.Ochodnicka E., Ochdnicky M., Belej K., Fusekova E. and Boselova L.

    (1995) Quantitative analysis of myelinated nerve fibers of periph-

    eral nerve in streptozotocin-induced diabetes mellitus. Mol. Chem.

    Neuropathol. 25, 225233.

    Oliver D., Lien C. C., Soom M., Baukrowitz T., Jonas P. and Fakler B.

    (2004) Functional conversion between A-type and delayed rectifier

    K+ channels by membrane lipids. Science 304, 265270.

    Park S. Y., Choi J. Y., Kim R. U., Lee Y. S., Cho H. J. and Kim D. S.

    (2003) Downregulation of voltage-gated potassium channel alpha

    gene expression by axotomy and neurotrophins in rat dorsal root

    ganglia. Mol. Cells 16, 256259.

    Park K. S., Mohapatra D. P., Misonou H. and Trimmer J. S. (2006)

    Graded regulation of the Kv2.1 potassium channel by variable

    phosphorylation. Science 313, 976979.

    Pongs O. (1999) Voltage-gated potassium channels: from hyperexcit-ability to excitement. FEBS Lett. 452, 3135.

    Rasband M. N., Park E. W., Vanderah T. W., Lai J., Porreca F. and

    Trimmer J. S. (2001) Distinct potassium channels on pain-sensing

    neurons. Proc. Natl Acad. Sci. USA 98, 1337313378.

    Robertson B. E. and Nelson M. T. (1994) Aminopyridine inhibition and

    voltage dependence of K+ currents in smooth muscle cells from

    cerebral arteries. Am. J. Physiol. 267, C1589C1597.

    Safronov B. V., Bischoff U. and Vogel W.. (1996) Single voltage-gated

    K+ channels and their functions in small dorsal root ganglion

    neurones of rat. J. Physiol. 493 (Pt 2), 393408.

    Stuhmer W., Ruppersberg J. P., Schroter K. H., Sakmann B., Stocker M.,

    Giese K. P., Perschke A., Baumann A. and Pongs O. (1989)

    Molecular basis of functional diversity of voltage-gated potassium

    channels in mammalian brain. EMBO J. 8, 32353244.

    Tapley P., Lamballe F. and Barbacid M. (1992) K252a is a selective

    inhibitor of the tyrosine protein kinase activity of the trk

    family of oncogenes and neurotrophin receptors. Oncogene 7,

    371381.Thompson S. W., Bennett D. L., Kerr B. J., Bradbury E. J. and

    McMahon S. B. (1999) Brain-derived neurotrophic factor is an

    endogenous modulator of nociceptive responses in the spinal cord.

    Proc. Natl Acad. Sci. USA 96, 77147718.

    Tonra J. R., Curtis R., Wong V., Cliffer K. D., Park J. S., Timmes A.,

    Nguyen T., Lindsay R. M ., Ac heson A. and D iStefano P. S . (19 98)

    Axotomy upregulates the anterograde transport and expression of

    brain-derived neurotrophic factor by sensory neurons. J. Neurosci.

    18, 43744383.

    Uchida H., Sasaki K., Ma L. and Ueda H. (2010) Neuron-restrictive

    silencer factor causes epigenetic silencing of Kv4.3 gene after

    peripheral nerve injury. Neuroscience 166, 14.

    Vydyanathan A., Wu Z. Z., Chen S. R. and Pan H. L. (2005) A-type

    voltage-gated K+ currents influence firing properties of isolectin

    B4-positive but not isolectin B4-negative primary sensory neurons.J. Neurophysiol. 93, 34013409.

    Wetmore C. and Olson L. (1995) Neuronal and nonneuronal expression

    of neurotrophins and their receptors in sensory and sympathetic

    ganglia suggest new intercellular trophic interactions. J. Comp.

    Neurol. 353, 143159.

    Wu Z. Z., Li D. P., Chen S. R. and Pan H. L. (2009) Aminopyridines

    potentiate synaptic and neuromuscular transmission by targeting

    the voltage-activated calcium channel beta subunit. J. Biol. Chem.

    284, 3645336461.

    Yang E. K., Takimoto K., Hayashi Y., de Groat W. C. and Yoshimura N.

    (2004) Altered expression of potassium channel subunit mRNA

    and alpha-dendrotoxin sensitivity of potassium currents in rat

    dorsal root ganglion neurons after axotomy. Neuroscience 123,

    867874.

    Youssoufian M. and Walmsley B. (2007) Brain-derived neurotrophicfactor modulates cell excitability in the mouse medial nucleus of

    the trapezoid body. Eur. J. Neurosci. 25, 16471652.

    Zhou X. F. and Rush R. A. (1996) Endogenous brain-derived neuro-

    trophic factor is anterogradely transported in primary sensory

    neurons. Neuroscience 74, 945953.

    Zhou X. F., Chie E. T., Deng Y. S., Zhong J. H., Xue Q., Rush R. A. and

    Xian C. J. (1999) Injured primary sensory neurons switch pheno-

    type for brain-derived neurotrophic factor in the rat. Neuroscience

    92, 841853.

    Zurek J. R., Nadeson R. and Goodchild C. S. (2001) Spinal and sup-

    raspinal components of opioid antinociception in streptozotocin

    induced diabetic neuropathy in rats. Pain 90, 5763.

    2010 The Authors

    Journal Compilation 2010 International Society for Neurochemistry J Neurochem (2010) 114 14601475

    BDNF and K+ channels in diabetic neuropathy | 1475