Skip to main content

ORIGINAL RESEARCH article

Front. Cell. Neurosci., 30 August 2013
Sec. Cellular Neurophysiology
Volume 7 - 2013 | https://doi.org/10.3389/fncel.2013.00137

β1- and β3- voltage-gated sodium channel subunits modulate cell surface expression and glycosylation of Nav1.7 in HEK293 cells

  • 1Pain Center, Department of Anesthesiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
  • 2Department of Clinical Research, University of Bern, Bern, Switzerland
  • 3Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland

Voltage-gated sodium channels (Navs) are glycoproteins composed of a pore-forming α-subunit and associated β-subunits that regulate Nav α-subunit plasma membrane density and biophysical properties. Glycosylation of the Nav α-subunit also directly affects Navs gating. β-subunits and glycosylation thus comodulate Nav α-subunit gating. We hypothesized that β-subunits could directly influence α-subunit glycosylation. Whole-cell patch clamp of HEK293 cells revealed that both β1- and β3-subunits coexpression shifted V½ of steady-state activation and inactivation and increased Nav1.7-mediated INa density. Biotinylation of cell surface proteins, combined with the use of deglycosydases, confirmed that Nav1.7 α-subunits exist in multiple glycosylated states. The α-subunit intracellular fraction was found in a core-glycosylated state, migrating at ~250 kDa. At the plasma membrane, in addition to the core-glycosylated form, a fully glycosylated form of Nav1.7 (~280 kDa) was observed. This higher band shifted to an intermediate band (~260 kDa) when β1-subunits were coexpressed, suggesting that the β1-subunit promotes an alternative glycosylated form of Nav1.7. Furthermore, the β1-subunit increased the expression of this alternative glycosylated form and the β3-subunit increased the expression of the core-glycosylated form of Nav1.7. This study describes a novel role for β1- and β3-subunits in the modulation of Nav1.7 α-subunit glycosylation and cell surface expression.

Introduction

Voltage-gated sodium channels (Navs) are large glycoprotein complexes responsible for the initial rising phase of the action potential in excitable cells. They are composed of a highly processed α-subunit and are associated to one or more β-subunits (Brackenbury and Isom, 2011). The α-subunit is the pore-forming unit of the channel through which the Na+ ions pass (Catterall, 2000). Nine genes encoding Nav α-subunits have been found in the human genome. In addition, four genes coding for the different Nav β-subunits have been identified: SCN1B (Isom et al., 1992; Kazen-Gillespie et al., 2000), SCN2B (Isom et al., 1995a), SCN3B (Morgan et al., 2000) and SCN4B (Yu et al., 2003) coding for β1- to β4-subunits, respectively. The α-subunit is composed of four homologous domains (Noda et al., 1984). Each of these domains contains six α-helical transmembrane domains (S1–S6). S1–S4 form the voltage-sensing domains and thus regulate α-subunit opening. S5 and S6 form the pore of the channel (Guy and Seetharamulu, 1986; Payandeh et al., 2011). The pore-forming α-subunit permits the flow of Na+, but its biophysical properties are modulated by the β-subunits (Isom et al., 1995b), most likely via direct interference with gating (Zimmer and Benndorf, 2002). The influence of β-subunits on the biophysical properties of the recorded sodium current (INa) vary with cell type, possibly due to different endogenous β-subunit expression and the presence of different partner proteins (Meadows and Isom, 2005). The β-subunits also participate in cell–cell adhesion and cell migration via the interaction with the extracellular matrix and cytoskeletal molecules. They also serve as important signaling molecules (Isom, 2001). Naturally occurring genetic variants in humans and genetically modified animal models have shown that β-subunits are implicated in numerous diseases, i.e., pain, epilepsy, migraines and cardiac arrhythmias (Brackenbury and Isom, 2011). This highlights their importance in the regulation of cellular excitability. The β-subunits are composed of an extracellular immunoglobulin-like domain in the N-terminal region, a single transmembrane segment and an intracellular carboxy-terminus tail (Isom et al., 1992). β1- and β3-subunits interact with the α-subunit via a non-covalent bond (Hartshorne et al., 1982), while β2- and β4-subunits are covalently linked to the α-subunit via disulfide bonds (Hartshorne et al., 1982; Messner and Catterall, 1985; Yu et al., 2003).

Out of the total pool of Navs, most of the α-subunits are localized intracellularly: in the endoplasmic reticulum (ER) for synthesis, in the Golgi where post-translational modifications occur and in the secretory pathway where they are trafficked to the plasma membrane to exert their main functions (Schmidt et al., 1985; Ritchie et al., 1990; Okuse et al., 2002). In the ER and the Golgi, Nav α-subunits undergo extensive sequential glycosylation (Waechter et al., 1983; Schmidt and Catterall, 1987), a process involving the addition of N-acetylglucosamine capped by sialic acid residues and the sequential addition of oligosaccharide chains. Glycosylation can account for up to 30% of the α-subunit molecular weight (Messner and Catterall, 1985). Protein glycosylation serves various functions such as protein folding, cell signaling, protection from proteases, cell-cell adhesion and regulation. It has also been implicated in development and immunity (Moremen et al., 2012). Glycosylation modifies the gating properties of the Nav α-subunits (Recio-Pinto et al., 1990; Bennett et al., 1997; Zhang et al., 1999; Tyrrell et al., 2001), most likely by interfering with the electric field near the gating sensors (Bennett et al., 1997; Cronin et al., 2005; Ednie and Bennett, 2012).

Because both β-subunits and glycosylation modify the intrinsic biophysical properties of the Nav α-subunit, we hypothesized that β-subunits might directly influence α-subunit glycosylation. This study investigated the effect of the four β-subunits on the Nav1.7-mediated current when co-expressed in HEK293 cells. Each of the four β-subunits influenced the biophysical properties and kinetics of the Nav1.7-mediated current to varying degrees, but only the β1- and β3-subunits increased Nav1.7 current density. Cell surface biotinylation and subsequent deglycosylation of the samples revealed the presence of differentially glycosylated forms of Nav1.7 in the cell; a core-glycosylated and a fully-glycosylated form of Nav1.7. β1- and β3-subunits mediated the differentially glycosylated form of Nav1.7 and enhanced its expression at the membrane. This suggests that the increase in Nav1.7 INa may be explained by a glycosylation-dependent stabilization of Nav1.7 at the cell membrane. This work reveals a novel mechanism by which Nav β-subunits modulate α-subunit glycosylation and cell surface density.

Materials and Methods

DNA Constructs

Nav1.7, β1-, β2-, and β4-subunit cDNA cloned into pCINh and β3-subunit cloned into pFBM were provided by Dr. S. Tate (Convergence Pharmaceuticals, Cambridge, UK).

Cell Culture and Transfection

Human embryonic kidney (HEK293) cells were cultured in DMEM medium supplemented with 10% FBS, 4 mM Glutamine and 20 μg/ml Gentamicin, at 37°C in a 5% CO2 incubator (Life Technologies Inc.). For patch clamp experiments, 1 μ g of Nav1.7 cDNA concomitantly with 0.4 μ g of a β-subunit and 0.8 μ g EBO-pCD-Leu2-CD8 cDNA encoding CD8 antigen as a reporter gene were transfected using the Ca2+-phosphate method in a T25 (~2 × 106 cells). For biotinylation and deglycosylation assays, HEK293 cells were transiently co-transfected with 6 μ g of Nav1.7 and 6 μ g of each β-subunit or empty vector mixed with 30 μ l JetPEI (Polyplus-Transfection) and 250 μ l 150 mM NaCl in a P100 dish (~9 × 106 cells, BD Falcon). For a negative control, the cells were transfected with 12 μ g of empty vector. The cells were used in patch clamp or biochemical experiments 48 h post transfection.

Cell Surface Biotinylation Assay

HEK293 cells transiently co-transfected were treated with 0.5 mg/ml EZ-link™ Sulfo-NHS-SS-Biotin (Thermo Scientific) in cold 1X PBS for 15 min at 4°C. The cells were then washed twice with 200 mM Glycine in cold 1X PBS to inactivate biotin, and twice with cold 1X PBS to remove excess biotin. The cells were then lysed with 1X lysis buffer [50 mM HEPES pH 7.4; 150 mM NaCl; 1.5 mM MgCl2; 1 mM EGTA pH 8; 10% Glycerol; 1% Triton X-100; 1X Complete Protease Inhibitor Cocktail (Roche)] for 1 h at 4°C. Whole cell lysates were centrifuged at 16,000 g at 4°C for 15 min. 2 mg of the supernatant was incubated with 50 μ l Streptavidin Sepharose High Performance beads (GE Healthcare) for 2 h at 4°C, and the remaining supernatant was kept as input. The beads were subsequently washed five times with 1X lysis buffer before elution with 50 μ l of 2X NuPAGE sample buffer (Invitrogen) and 100 mM DTT at 37°C for 30 min. These biotinylated fractions were analyzed as Nav1.7 expression at the cell surface. The input fractions, representing total expression of Nav1.7, were resuspended with 4X NuPAGE sample buffer plus 100 mM DTT to give a concentration of 1 mg/ml (60 μ g/well) and were then incubated at 37°C for 30 min.

Deglycosylation Assay

For the total fractions, 60 μ g of proteins of whole cell lysates were denatured at 37°C for 30 min in the presence of 1X Glycoprotein denaturing buffer. The denatured protein lysates were subsequently incubated at 37°C for 1 h with 1500 units PNGaseF (New England Biolabs) in the presence of 1X NP-40 and 1X G7 buffer to cleave most of the high mannose, hybrid and complex oligosaccharides from N-linked glycoproteins. The reaction was stopped by adding 4X NuPAGE sample buffer plus 100 mM DTT and incubating them at 37°C for 30 min. For the biotinylated fractions, 35 μ l ddH2O were added into Streptavidin Sepharose High Performance beads previously incubated with whole cell lysate and denatured at 37°C for 30 min in the presence of 1X glycoprotein denaturing buffer. The denatured proteins bound to Streptavidin beads were subsequently incubated at 37°C for 1 h with 2000 units of PNGaseF in the presence of 1X NP-40 and 1X G7 buffer. Following this incubation step, the beads were washed five times with the same lysis buffer used in the biotinylation assay and eluted with 2X NuPAGE sample buffer and 100 mM DTT at 37°C for 30 min.

Western Blots

Protein samples were separated on a 5–15% polyacrylamide gradient gel and blotted onto a nitrocellulose membrane using TransBlot Turbo transfer system (Biorad, Hercules). Antibody detections were performed in the SNAP i.d. system (Millipore) using the following antibodies: mouse monoclonal anti-Nav1.7 clone N68/6 (UC Davis/National Institute of Health (NIH) NeuroMab Facility, University of California), mouse monoclonal clone 464.6 anti-Na+/K+ ATPase α-1 (Abcam), rabbit polyclonal anti-actin A2066 (Sigma) and rabbit polyclonal anti-β4 (EnoGene). Rabbit polyclonal homemade anti-β1, anti-β2 and anti-β3 antibodies were provided by Dr. S. Tate (Convergence Pharmaceuticals, Cambridge, UK). Infrared IRDyeTM (680 or 800 CW)-linked goat anti-rabbit or anti-mouse IgG (LI-COR Biosciences) was used as secondary antibody. The blots were revealed and quantified with Odyssey Li-Cor (Lincoln).

Electrophysiology

Twenty-four hours after transfection, cells were split at low density and whole-cell recordings were performed 48 h after transfection. Anti-CD8 beads (Dynal, Oslo, Norway) were used to identify transfected cells. Whole cell patch-clamp recordings were carried-out using an internal solution containing 60 mM CsCl, 70 mM Cs Aspartate, 11 mM EGTA, 1 mM MgCl2, 1 mM CaCl2, 10 mM HEPES, and 5 mM Na2-ATP, pH 7.2 with CsOH and an external solution containing 130 mM NaCl, 2 mM CaCl2, 1.2 mM MgCl2, 5 mM CsCl, 10 mM HEPES, 5 mM glucose, pH 7.4 with CsOH. Data were recorded with a VE-2 amplifier (Alembic Instruments, Montreal, Canada) or an Axon amplifier 700A and analyzed using pClamp software (version 8, Molecular Devices), Kaleidagraph (version 4.03) and MatLab. The sampling interval was set to 5 μ s (200 kHz) and low-pass filtering to 5.0 kHz. Resistance of the borosilicate pipettes (World Precision Instruments, Sarasota, FL, USA) was 2–6 MΩ. Leakage current was subtracted using the P/4 procedure. INa densities (pA/pF) were obtained by dividing the peak INa by the cell capacitance obtained from the pClamp function. Voltage dependence of activation (SSA) curves were determined from I/V curves where the Na+ current was evoked from a holding potential of −100 mV to test pulses of 100 ms ranging from −120 to +30 mV in increments of 5 mV. The linear ascending segment of the I/V relationship was used to estimate the reversal potential for each trace. Time constant of inactivation was determined by fitting the current decay with the Levenberg-Marquardt single exponential function. The time constant was plotted against the test voltage, with I = A * exp (−t/τ) + C: where I is the current, A is the percentage of channel inactivation with the time constant τ, t is time and C if the steady-state asymptote. Steady-state inactivation curves (SSI) were measured from a holding potential of −120 mV using 500 ms prepulses to the indicated potentials, followed by a test pulse to 0 mV. To quantify the voltage-dependence of SSA and SSI, data from individual cells were fitted with the Boltzmann relationship, y (Vm) = 1/(1 + exp[(VmV½)/k]), in which y is the normalized current or conductance, Vm is the membrane potential, V½ is the voltage at which half of the available channels are inactivated and k is the slope factor.

Recovery from inactivation curves (RFI or “repriming”) were obtained with a standard two-pulse protocol consisting of a depolarizing pulse from a holding potential of −120 to 0 mV for 50 ms to inactivate the channels, followed by a variable duration (from 0.5 to 3000 ms) step back to −120 mV to promote recovery. Channel availability was assessed with the first standard test pulse at 0 mV. The normalized currents of the second pulse at 0 mV were plotted against the recovery interval. We calculated t½ (ms), the time necessary for half of the channels to recover from the first pulse, by interpolation from a linear relation between the 2 points juxtaposing half recovery (y1 < 0.5 < y2), using the equation x = [0.5–(y1 x2y2 x1)/(x2x1)]*(x2x1)/(y2y1).

Quantitative Real-Time Reverse Transcription PCR (qRT-PCR)

HEK293 cells transfected with Nav1.7 (1 μ g) alone or with each of β-subunits (0.4 μ g) were collected in RNA-later solution (Qiagen, Basel, Switzerland). mRNA was extracted and purified with RNeasy Plus Mini kit (Qiagen) and quantified using RNA 6000 Nano Assay (Agilent Technologies AG, Basel, Switzerland). A total of 600 ng of RNA was reverse transcribed for each sample using Omniscript reverse transcriptase (Qiagen). Nav1.7 primer's sequence is as follow; 5′-TCTGTCTGAGTGTGTTTGCACTAA-3′ and 5′-AAGTCTTCTTCACTCTCTAGGGTATTC-3′. We used GAPDH as reference gene to normalize Nav1.7 mRNA expression. Gene-specific mRNA analyses were performed using the iQ SYBR-green Supermix (BioRad, Reinach, Switzerland) and the iQ5 real-time PCR detection system (BioRad). Only reactions with appropriate amplification and melting curves determining the amplicon specificity were analyzed. For all conditions tested we used n = 3 samples. All samples were run in triplicate.

Statistics

For electrophysiological experiments (current densities and biophysical properties) normality with D'Agostino-Pearson was tested to determine whether a regular One-Way ANOVA and post-hoc Bonferroni tests, or the non-parametric equivalent test (Kruskal-Wallis test and Dunn post-hoc tests), should be performed. For RFI, a Two-Way ANOVA was used to compare Nav1.7 alone with Nav1.7 co-transfected with each β-subunit, and the impact of the voltage on this comparison. Biochemical experiments and transcriptional quantification data were analyzed using bilateral Student's t.

Results

The functional impact of the co-expression of the four β-subunits on Nav1.7-mediated INa was studied by performing whole cell patch-clamp experiments in HEK293 cells. Each β-subunit was independently co-transfected with Nav1.7 and then compared to Nav1.7 expressed alone. Figure 1A shows typical traces of Nav1.7 INa obtained with a current-voltage protocol. A hastening of the Nav1.7 current decay kinetics was observed with each of the β-subunits tested (Figure 1B). The shortening of the Nav1.7 time constant of current decay was observed for a wide range of voltages and showed voltage-dependency for every β-subunit (Figure 1B). The shortening was particularly prominent for the β3-subunit. In addition, β1- and β3-subunits also significantly increased (~2-fold) Nav1.7-mediated current density as compared to Nav1.7 alone or to Nav1.7 co-expressed with β2- or β4-subunits (Figure 1C and Table 1). We also observed that β2 and β4-subunits did not antagonize β1 and β3-subunits-dependent up-regulation, and that the two latter have additive positive effect on Nav1.7-mediated current (data not shown).

FIGURE 1
www.frontiersin.org

Figure 1. β-subunits regulate Nav1.7 currents. (A) Typical whole-cell Na+ currents of HEK293 cells transfected with Nav1.7 alone or Nav1.7 co-expressed with individual β-subunits elicited with a typical current-voltage protocol. (B) Voltage-dependence of current decay of Nav1.7 alone compared to Nav1.7 with each individual β-subunit. Inset: Normalized representative current traces of Nav1.7 elicited by test pulses at 0 mV. Co-transfection of the β1- (p = 0.011, n = 39), β2- (p < 0.0001, n = 25), β3- (p < 0.0001, n = 16), and β4-subunit (p = 0.006, n = 13) decreased the time constant decay as compared to Nav1.7 alone (n = 81). Two-Ways ANOVA and Bonferroni post-hoc tests. Data are expressed as mean ± s.e.m. (C) INa densities from HEK293 cells transfected with Nav1.7 alone or co-transfected with individual β-subunits. β1- (n = 55) and β3-subunits (n = 34), but not β2- (n = 67) nor β4-subunits (n = 27), increased the Nav1.7 current densities. p < 0.0001 for β1- and β3-subunits with One-Way ANOVA followed by Bonferroni post-hoc tests. Data are expressed as mean ± s.e.m. and were normalized to Nav1.7 alone for each experiments. Values can be found in Table 1. *p < 0.05, **p < 0.01 and ***p < 0.001.

TABLE 1
www.frontiersin.org

Table 1. Biophysical properties of Nav1.7 alone or upon β-subunit co-transfection in HEK293 cells.

Whether the INa density increase mediated by both β1- and β3-subunits was also accompanied by alterations of other Nav1.7 biophysical properties was also assessed. The voltage dependence of macroscopic INa activation and inactivation (see Materials and Methods) of Nav1.7 in the absence and presence of each β-subunit was recorded and analyzed. The co-transfection of the β1-subunit significantly shifted the V½ of steady-state inactivation toward depolarized potentials by ~5.8 mV, but had no influence on V½ of activation (Figure 2A and Table 1). The β3-subunit shifted the V½ of inactivation toward depolarized potentials by ~3.5 mV and the V½ of activation toward hyperpolarized potentials by ~3.7 mV (Figure 2C and Table 1). Neither the β2- nor β4-subunits affected Nav1.7 voltage dependence of activation or inactivation (Figures 2B,D and Table 1).

FIGURE 2
www.frontiersin.org

Figure 2. The influence of β1- and β3-subunits on Nav1.7 voltage-dependence of activation and inactivation. (A–D) Normalized currents for both activation and steady-state inactivation (see Materials and Methods) are plotted against the test potential. Each panel compares Nav1.7 alone to Nav1.7 co-expressed with individual β-subunits. A minor but significant effect on the V½of activation was observed for β3-subunit (n = 17, hyperpolarizing shift, p < 0.05) co-expression as compared to Nav1.7 alone (n = 81). β1- (n = 39), β2- (n = 25) and β4-subunits (n = 13) did not modify the V½of activation of Nav1.7. The effect on the V½ of inactivation was highly significant for β1- (n = 51, depolarizing shift with p < 0.0001) and β3-subunit (n = 18, depolarizing shift with p < 0.0001) compared to Nav1.7 alone (n = 92); whereas β2- (n = 43) and β4-subunits (n = 20) had no effect. Individual points are the mean ± s.e.m. of the normalized current at each voltage point. The smooth curves are Boltzmann fits whose equations give both the V½of activation and inactivation (midpoints) and their associated slope factors (see Materials and Methods). The V½ of steady-state activation and inactivation comparing Nav1.7 alone vs. the co-expression with each subunit were tested by One-Way ANOVA followed by Bonferroni's multiple comparison tests. Values and statistics can be found in Table 1.

The influence of the β-subunits on recovery from inactivation (RFI) was also tested. Because the RFI relationships could not always be fitted with the exponential functions to the same degree, an interpolation from a linear relation between the 2 points juxtaposing half recovery to obtain the half-time (t½) of RFI was used. Only the β1-subunit significantly hastened t½ of RFI to 6.19 vs. 7.55 ms for the control (Figures 3A–D and Table 1).

FIGURE 3
www.frontiersin.org

Figure 3. Only β1-subunit significantly increases Nav1.7 recovery from inactivation (RFI). (A–D) RFI from HEK293 cells transfected with Nav1.7 alone or co-transfected with individual β-subunits. Smooth curves were fitted incorporating a geometric weight to arrive at the final curve (no equation) and the t½ was calculated by interpolation on the x-axis from a linear relation between the 2 points juxtaposing half recovery (y1 < 0.5 < y2, see Materials and Methods). Only when co-expressed with β1-subunit (n = 27, p < 0.01) was Nav1.7 RFI significantly faster as compared to Nav1.7 alone (n = 72). β2- (n = 34), β3- (n = 16), and β4-subunits (n = 19) did not significantly alter RFI when co-expressed with Nav1.7. Individual points are the mean ± s.e.m. of the normalized current at each time point. Non-parametric One-Way analysis of variance (Kruskal-Wallis test) with Dunn post-hoc tests to compare each subunit co-expressed with Nav1.7 vs. Nav1.7 alone.

As only minor modifications of the INa biophysical properties were observed, it is unlikely that the 2-fold increase in the Nav1.7 current density mediated by β1- and β3-subunits is only due to alterations of the single channel properties. Whether the increase of the Nav1.7 current may have been due to an increase in channel synthesis was investigated. Nav1.7 mRNA levels remained unchanged with β-subunit co-transfection, as observed by q-RT-PCR (Figure 4), discounting this hypothesis.

FIGURE 4
www.frontiersin.org

Figure 4. Nav1.7 transcripts upon β-subunit co-transfection. Bar graph showing transcriptional levels of Nav1.7 in control conditions (co-transfected with empty vector) over Nav1.7 levels when cells were co-transfected with each individual β-subunit. Transcripts were normalized to GAPDH and run in triplicate. Data represent the mean ± s.e.m., n = 3 independent transfections for every condition.

Whether the Nav1.7-mediated INa upregulation could be due to an increase of Nav1.7 protein density at the cell membrane was investigated by performing biotinylation of plasma membrane proteins. After lysis, proteins were sampled under reducing conditions known to dissociate the covalently bound β2- and β4-subunits from α-subunits (Messner and Catterall, 1985). Co-transfection of β1-, β2-, and β4-subunits significantly decreased Nav1.7 protein expression in the total cell lysate fraction (input, Figure 5A). The quantification revealed a ~2-fold decrease for each of these three subunits. Co-transfection of the β3-subunit had no effect on Nav1.7 expression in the total cell lysate fraction. In the biotinylated membrane fraction two bands at different apparent molecular weights were observed when Nav1.7 was expressed alone (white and black arrow heads in Figure 5A). These bands correspond to different glycosylated states of Nav1.7 as demonstrated by using deglycosylating enzymes (Figure 5B). Endoglycosidase H (EndoH) only cleaves core N-glycans from proteins whereas Peptide-N-Glycosidase F (PNGaseF) does not discriminate between full and core glycosylated proteins. Of the two bands of biotinylated Nav1.7, only the lower was sensitive to EndoH and was shifted to an apparent lower molecular weight band (compare white arrowhead in the first lane to gray arrowhead in the third lane), indicating that this band corresponds to the core-glycosylated form of the channel (Figure 5B). Because PNGaseF was able to digest both bands, it can be proposed that the higher band corresponds to the fully-glycosylated form of Nav1.7. The lower band of biotinylated Nav1.7 migrates at the same apparent molecular weight as the band observed in the input fraction (white arrow heads in Figure 5A) suggesting that most of Nav1.7 in the intracellular pool is core-glycosylated. This is consistent with the channel being early and rapidly, but only partially, glycosylated after its synthesis. The upper band in the total cell lysate fraction was faint and blurry (Figure 5A), suggesting that the fully-glycosylated channel only represents a small fraction of the total Nav1.7 cellular pool. It was only by enriching the membrane proteins through the precipitation of the biotinylated membrane fraction (the ratio between the amount of lysate protein loaded and the amount of streptavidin beads needed to precipitate biotinylated proteins was ~1:30) that the upper band was distinctly observed. Co-expression of the β1-subunit reproducibly shifted the upper band to an intermediate migrating band of lower apparent molecular weight. This suggests that the β1-subunit mediates an alternative glycosylated form of Nav1.7. When comparing the β1-subunit-modified intermediate band with the upper band of the control condition (Nav1.7 alone), a significant increase in signal intensity was observed (Figure 5A, quantification), which is consistent with the increase in the Nav1.7 current density (Figure 1C). Co-transfection of the β2-subunit neither modified the glycosylation pattern nor the expression of any of the two bands, consistent with the fact that the current density was not modified. β3-subunit expression also altered the Nav1.7 band pattern in the biotinylated fractions. The upper band overlapped with the lower band under the migrating conditions used. The β3-subunit significantly increased (~7-fold) the intensity of the lower band as compared to the lower band of control, consistent with the increase of the Nav1.7 current density elicited by the β3-subunit (Figure 1C). Finally, β4-subunit co-transfection led to a small but significant decrease of the lower band.

FIGURE 5
www.frontiersin.org

Figure 5. β1- and β3-subunit mediate differential forms of Nav1.7 whose expression is increased at the membrane. (A) Representative western blot of a biotinylation assay with total lysate (input, left) and cell surface (biotinylation, right) fractions from HEK293 cells transiently transfected with Nav1.7 alone, or co-expressed with each individual β-subunit and the associated quantifications. Input: Nav1.7 is detected in two forms: a fast migrating band (~250 kDa, that will be referred to as lower band) that consist mostly of the Nav1.7 immunoreactive signal and a slow migrating band (~280 kDa, that will be referred to as upper band). β1- (p = 0.006), β2- (p = 0.003), and β4-subunits (p = 0.009) significantly decreased Nav1.7 expression, whereas the β3-subunit had no effect (p = 0.570). Because the upper band was below the sensitivity threshold, both bands were quantified together. Biotinylation: Nav1.7 membrane protein is detected in three forms. When expressed alone, one lower band (white triangle, ~250 kDa) and one upper band (black triangle, ~280 kDa) were present (for identification of these bands, see Panel B). When the β1-subunit is co-expressed, the upper band was clearly shifted into an intermediate migrating band (~260 kDa) with increased expression (p = 0.047). β2- and β4-subunits revealed the same pattern as when Nav1.7 was transfected alone and did not change its expression, except for the small decrease of the lower band when the β4-subunit is co-transfected (p = 0.020). The β3-subunit clearly increased Nav1.7 immunoreactivity of the lower band (p < 0.0001). For input and biotinylation fractions, actin and the α1-subunit of NaK-ATPase were used as biotin leakiness and loading controls, respectively. Data represent mean ± s.e.m, n = 4 independent experiments. Student's unpaired t-test, each condition being compared with Nav1.7. *p < 0.05, **p < 0.01 and ***p < 0.001. (B) Representative western blot and identification of glycosylation state of Nav1.7 in biotinylated fraction from HEK293 cells transiently transfected with Nav1.7. EndoH only cleaves the lower band of biotinylated Nav1.7, demonstrating that this band represents the core-glycosylated form of the channel. The upper band is digested by PNGaseF, demonstrating that it corresponds to fully-glycosylated form of the channel. PNGaseF can also digest the core-glycosylated form of Nav1.7.

To confirm that the different bands observed when β-subunits are coexpressed, particularly β1 and β3-subunits, represent alternative glycosylated form of Nav1.7, we again incubated the input and biotinylated fractions with PNGaseF. A small but consistent shift of the Nav1.7 band into a lower apparent molecular weight band in the total cell lysate fraction was observed (the white arrow heads shifted to the gray arrow heads, Figure 6). Furthermore, when incubating the biotinylated fraction of β-subunit and Nav1.7 co-expression experiments with PNGaseF, all of the Nav1.7 bands shifted to a single band (gray arrow heads) of the same molecular weight. This confirms that the β1- and β3-subunits modulate differential glycosylation patterns on Nav1.7 (Figure 6, black and white arrow heads). Noteworthy, when comparing the single band of Nav1.7 when samples are treated with PNGaseF in the input fraction with the one in the biotinylated fraction, it seems that this band migrates slower in the input as compared to biotinylated fraction when β-subunits are coexpressed (compare bands highlighted by gray arrows for each blots). This may be due to other post-translational modification such as sialylation or palmitoylation of the channel. Further experiments using desialylation or depalmitoylation treatment are needed to confirm this possibility.

FIGURE 6
www.frontiersin.org

Figure 6. The different forms of Nav1.7 observed with β-subunits are due to differential glycosylation patterns. Western blot of a biotinylation assay followed by deglycosylation with total lysate and cell surface fractions from HEK293 cells transiently transfected with Nav1.7 alone, or co-expressed with each individual β-subunit. Samples were non-treated or treated with Peptide: N-Glycosidase F (PNGaseF) to remove glycosylated residues of the protein. The total lysate Nav1.7 band (black/white triangle) was slightly shifted to an apparent lower molecular weight (gray triangle) when treated with PNGaseF. In the biotinylation fraction, the pattern of Nav1.7 glycosylation by the β-subunits was the same as in Figure 4.(black and white triangles). When treated with PNGaseF, all the different bands shifted to the lower band of the same apparent molecular weight, irrespective of the β-subunit co-expressed.

Discussion

This study demonstrates that Nav β1- and β3-subunits modulate the cell surface expression and glycosylation patterns of Nav1.7 when co-expressed in HEK293 cells. It also confirms that the β-subunits differentially modulate the biophysical properties of Nav1.7.

The observation that β1- and β3-subunits strongly increased Nav1.7 INa density contrasts with several recent studies. The study performed by Ho et al. (2012) showed no impact of any of the β-subunits on Nav1.7 current density in HEK293 cells. The other studies showed no impact using different cell expression systems (Sangameswaran et al., 1997; Morgan et al., 2000; Vijayaragavan et al., 2001, 2004). These discrepant observations underline the influence of the cellular background when studying Navs α-subunit regulation by β-subunits. Even though this study and the one by Ho et al. (2012) both used HEK293 cells, Ho et al., used a clonal cell line stably expressing rat Nav1.7 cDNA, whereas the present study used transiently transfected human Nav1.7 cDNA. In a stable cell line, it is likely that a significant fraction of α-subunits are already anchored at the plasma membrane; thus it is possible that the ones interacting with the transfected β-subunits only reflect a small fraction of membrane Navs α-subunits. Under the conditions of transient transfection, all the membrane Navs α-subunits are synthesised de novo and are thus more likely to interact with β-subunits. It is also possible that other differences, such as the origin of the HEK293 cells or even passage numbers, may play a significant role in the observed effects. Other studies have shown, however, that β1- and β3-subunits can increase the current density of several other Nav isoforms (Nuss et al., 1995; Smith and Goldin, 1998; Fahmi et al., 2001; Zimmer and Benndorf, 2002).

A significant shortening of the time constant of current decay was observed when Nav1.7 was co-expressed with each individual subunit. A rapid rate of inactivation tends to reduce the refractory period of Nav1.7, meaning that β-subunits can enhance cell excitability via this mechanism. The hastening of RFI by the β1-subunit might also reduce the duration of the refractory periods, allowing for faster repetitive firing of neurons.

The β1-subunit shifted V½ of inactivation toward more depolarized potentials, which should increase the number of channels available for opening in response to depolarization at a given voltage near the resting membrane potential (approximately −60 to −70 mV). The β3-subunit similarly influenced this parameter, and also shifted the V½ of activation toward hyperpolarized potentials, rendering the channel more likely to open at hyperpolarized voltages. These results are consistent with the findings of Ho et al., and account for the shift toward a hyperexcitable state.

The fact that the β1- and β3-subunits strongly increased Nav1.7 current density, but only modestly influenced the biophysical properties, suggests that the single-channel conductance is not altered. In line with the previous, single-channel recordings revealed that β1-subunit did not change the Nav1.5 open probability despite an important increase in current density (Nuss et al., 1995). As a consequence, it was hypothesized that these two subunits also increase Nav1.7 channel density at the cell surface. Biotinylation of cell surface proteins was performed and a strong decrease of the Nav1.7 signal in the input fraction when β1-, β2-, and β4-subunits were co-transfected was observed. This decrease was in contrast with the increase or lack of modification of the Nav1.7 current density. The reason for this decrease remains to be identified, but one can speculate that the expression of the β-subunits may decrease the ratio of intracellular Navs to plasma membrane Navs by hastening forward trafficking and stabilizing the channel at the cell surface. Furthermore, one of the important functions of glycosylation is the proper folding and protection of proteins with respect to proteolysis (Parodi, 2000). The thus far not observed role of β-subunits in altering Nav1.7 glycosylation might influence the degradation by the proteasome, accounting for the decrease in input. The β-subunits have also been proposed to act as chaperon proteins (Valdivia et al., 2010), further supporting a potential effect on degradation by the proteasome.

The analysis of the Nav1.7 protein at the plasma membrane and the subsequent treatment with the deglycosylating enzyme led to three novel findings: (1) Under normal conditions, Nav1.7 is present in two glycosylated forms, a core-glycosylated form with a molecular weight of ~250 kDa and a fully-glycosylated form with a molecular weight of ~280 kDa; (2) The β1-subunit can further mediate a third and intermediate migrating band, which likely represents an alternative fully-glycosylated form of Nav1.7; and (3) the β1-subunit increases the membrane expression of this alternative fully-glycosylated form of Nav1.7; whereas the β3-subunit increases the membrane expression of the core-glycosylated form.

In studies using chimeras between the β1- and β3-subunits, it was proposed that different parts of these subunits were involved in the modulation of gating by direct interaction with the α-subunits (Zimmer and Benndorf, 2002). The observation in this study that the β1- and β3-subunits can mediate the differential glycosylation of Nav1.7, and despite the well-documented causal link demonstrating that differential glycosylation leads to a modification of Navs α-subunit gating (Bennett et al., 1997; Zhang et al., 1999; Tyrrell et al., 2001), does not allow us to conclude that the β-subunit-mediated glycosylation of the α-subunit is responsible for the modulation of Nav1.7 gating. Further studies to identify the potential glycosylation site of Nav1.7 are necessary to determine whether the effects of the β-subunits on the biophysical properties of Nav1.7 are also dependent on this mechanism. Nevertheless, the observation that the β1- and β3-subunits influence the gating properties of Nav1.7 and alter its glycosylation pattern, while the β2- and β4-subunits do not, supports this hypothesis.

It was recently proposed that only the upper band of biotinylated Nav1.7 may reflect the functional fully-glycosylated form of the channel, whereas the lower band represents an intermediate and/or immature glycosylated form of the channel which does not participate in Na+ conductance (Laedermann et al., 2013). The upregulation of expression of the core-glycosylated form of Nav1.7 when the β3-subunit is co-transfected, along with the associated 2-fold increase in the Nav1.7 current density, suggests that there is no such dichotomy and that the link between the glycosylation and functionality of the channel is more complex. It is possible that these distinctly glycosylated forms of Nav1.7 differentially participate in Na+ conductance, but their relative contribution to the overall sodium current has yet to be determined. For instance, quantification of the shifted upper band when the β1-subunit was present revealed an increased signal intensity of ~30%, which is less important than the 100% increase of INa measured using the patch clamp approach. This underlines that, in addition to an increased stabilization of the channel, modification of single channel conductance by the intermediate glycosylated form of Nav1.7 may also partially contribute to the functional 2-fold increase in INa. This point, in addition to the fact that β-subunits have the ability to shift bands from one glycosylated state to another, demonstrates that quantification of biotinylated proteins needs to be interpreted with caution. It cannot be excluded that due to these shifts, some bands might contaminate the signal of another band.

General kinetic models of biosynthesis (Schmidt and Catterall, 1987) have proposed that the β2-subunit interacts with the α-subunit right before anchoring at the membrane. The present findings are consistent with such a model since both β2- and β4-subunits did not influence Nav1.7 glycosylation, and most likely interacted after the Golgi network (Figure 7). The α−β1 complex was previously shown to associate in the ER, enhancing the trafficking to the plasma membrane (Zimmer et al., 2002). Another study demonstrated that β1- and β3-subunits increase the efficiency of channel trafficking from the ER to the plasma membrane (Fahmi et al., 2001). The β3-subunit has also been shown to mask the ER-retention signal (Zhang et al., 2008). The results of the present work confirm earlier demonstrated interactions between the β- and α-subunits, and suggest an additional function of both: β1- and β3-subunits interact with the α-subunit in the ER/Golgi, where they regulate the differential glycosylation of Nav α-subunits, which in turn modulates the stabilization of the channel at the cell membrane (Figure 7). The mechanisms underlying their interaction and mediation of α-subunit glycosylation, as well as the identification of other potential isoforms that would be subject to such regulation, remain to be investigated.

FIGURE 7
www.frontiersin.org

Figure 7. Proposed scheme of the different intracellular pathways of α-subunits depending on the presence of different β-subunits. After synthesis in the rough endoplasmic reticulum (RER), α-subunits are rapidly folded and undergo a first step of glycosylation in the smooth endoplasmic reticulum (SER). It is known that N-acetlyglucosamine and oligosaccharide chains are bound on Asp residues of the protein, a process known as core-glycosylation. Newly synthesized glycoproteins are then translocated into the Golgi network, where they are subject to a second step of more complex glycosylation, involving many different enzymes. Once matured, proteins eventually translocate to the plasma membrane. The present findings suggest that core-glycosylated proteins can also be found anchored at the membrane. By some yet undefined mechanism, the β1-subunit interferes with the second glycosylation step (***full-glycosylation on the scheme) which leads to a modification of the glycosylation pattern of the α-subunits. The β1-subunit enhances the core-glycosylated form of Nav1.7. This suggests that the β1- and β3-subunits already interact with the α-subunits before the step of full-glycosylation of the channel occurring in the Golgi network. By enhancing the differential glycosylation pattern of Nav1.7, it can be proposed that the β1- and β3-subunits promote stabilization of the channel at the plasma membrane. On the contrary, it is likely that the β2- and β4-subunits, which have no effect on the glycosylation nor the anchoring, only briefly interact with the α-subunits before translocation of the channel to the plasma membrane. For sake of simplicity, the shown glycosylation patterns are arbitrary. Nav1.7 is depicted as being “freely” expressed in ER/Golgi and membrane networks for easier interpretation of the scheme. However, Nav1.7 is embedded in the membranes of the different organelles.

Physiological Relevance

These results were obtained in cellular expression system. Whether such mechanisms also occur in native cells remain to be investigated. This study used the Nav1.7 isoform, an important contributor to pain processing (Lampert et al., 2010). The electrophysiological results show that β1- and β3-subunits are able to increase Nav1.7 excitability, as demonstrated by the increase in peak current, kinetics, voltage-availability and repriming rate. Nav1.7 is expressed in high levels in all types of sensory neurons (Ho and O'leary, 2011). β-subunits are also expressed in sensory neurons, but to a variable extent depending on cell type (Takahashi et al., 2003; Ho et al., 2012). Furthermore, β1- and β3-subunit expression has been reported to be increased in pathological pain (Shah et al., 2000; Coward et al., 2001), where they have been implicated in the generation of hyperexcitability. It remains to be determined if Nav function is mediated by an altered pattern of α-subunit glycosylation, which is an important regulatory process of excitability in dorsal root ganglia neurons (Tyrrell et al., 2001).

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We thank S. Tate and V. Morisset (Convergence Pharmaceuticals) for providing Nav1.7 cDNA cloned into pCIN5h and β1-, β2-, and β4-subunits cDNA cloned into pCINh, as well as β3-subunit cloned into pFBM and the polyclonal antibodies anti-β1-, anti-β2-, and anti-β3-subunit. The monoclonal antibody N68/3 (anti-Nav1.7) was developed by and/or obtained from the UC Davis/NIH NeuroMab Facility, supported by NIH grant U24NS050606 and maintained by the Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA 95616. This study was supported by grants from the Swiss National Science Foundation (31003A–124996 to Isabelle Decosterd and 310030B-135693 to Hugues Abriel), the Synapsis Foundation (to Isabelle Decosterd and Hugues Abriel), the European Society of Anesthesiology (to Isabelle Decosterd and Hugues Abriel) and the Lemanic Neuroscience Doctoral School Ph.D. Fellowship (to Cédric J. Laedermann). We thank Dr. A. Felley for her comments on the manuscript. We would like to thank Prof. C. Kern, head of the Anesthesiology Department at CHUV for his support.

Supplementary Material

The Supplementary Material for this article can be found online at: http://www.frontiersin.org/Cellular_Neuroscience/10.3389/fncel.2013.00137/abstract

References

Bennett, E., Urcan, M. S., Tinkle, S. S., Koszowski, A. G., and Levinson, S. R. (1997). Contribution of sialic acid to the voltage dependence of sodium channel gating. a possible electrostatic mechanism. J. Gen. Physiol. 109, 327–343. doi: 10.1085/jgp.109.3.327

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Brackenbury, W. J., and Isom, L. L. (2011). Na channel beta subunits: overachievers of the ion channel family. Front. Pharmacol. 2:53. doi: 10.3389/fphar.2011.00053

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Catterall, W. A. (2000). From ionic currents to molecular mechanisms: the structure and function of voltage-gated sodium channels. Neuron 26, 13–25. doi: 10.1016/S0896-6273(00)81133-2

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Coward, K., Jowett, A., Plumpton, C., Powell, A., Birch, R., Tate, S., et al. (2001). Sodium channel beta 1 and beta 2 subunits parallel SNS/PN3 alpha-subunit changes in injured human sensory neurons. Neuroreport 12, 483–488. doi: 10.1097/00001756-200103050-00012

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Cronin, N. B., O'reilly, A., Duclohier, H., and Wallace, B. A. (2005). Effects of deglycosylation of sodium channels on their structure and function. Biochemistry 44, 441–449. doi: 10.1021/bi048741q

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ednie, A. R., and Bennett, E. S. (2012). Modulation of voltage-gated ion channels by sialylation. Compr. Physiol. 2, 1269–1301. doi: 10.1002/cphy.c110044

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Fahmi, A. I., Patel, M., Stevens, E. B., Fowden, A. L., John, J. E., Lee, K., et al. (2001). The sodium channel -subunit SCN3b modulates the kinetics of SCN5a and is expressed heterogeneously in sheep heart. J. Physiol. 537, 693–700. doi: 10.1113/jphysiol.2001.012691

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Guy, H. R., and Seetharamulu, P. (1986). Molecular model of the action potential sodium channel. Proc. Natl. Acad. Sci. U.S.A. 83, 508–512. doi: 10.1073/pnas.83.2.508

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hartshorne, R. P., Messner, D. J., Coppersmith, J. C., and Catterall, W. A. (1982). The saxitoxin receptor of the sodium channel from rat brain. evidence for two nonidentical beta subunits. J. Biol. Chem. 257, 13888–13891.

Pubmed Abstract | Pubmed Full Text

Ho, C., and O'leary, M. E. (2011). Single-cell analysis of sodium channel expression in dorsal root ganglion neurons. Mol. Cell Neurosci. 46, 159–166. doi: 10.1016/j.mcn.2010.08.017

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ho, C., Zhao, J., Malinowski, S., Chahine, M., and O'leary, M. E. (2012). Differential expression of sodium channel beta subunits in dorsal root ganglion sensory neurons. J. Biol. Chem. 287, 15044–15053. doi: 10.1074/jbc.M111.333740

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Isom, L., De Jongh, K., Patton, D., Reber, B., Offord, J., Charbonneau, H., et al. (1992). Primary structure and functional expression of the beta 1 subunit of the rat brain sodium channel. Science 256, 839–842. doi: 10.1126/science.1375395

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Isom, L. L. (2001). Sodium channel beta subunits: anything but auxiliary. Neuroscientist 7, 42–54. doi: 10.1177/107385840100700108

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Isom, L. L., Ragsdale, D. S., De Jongh, K. S., Westenbroek, R. E., Reber, B. F. X., Scheuer, T., et al. (1995a). Structure and function of the β2 subunit of brain sodium channels, a transmembrane glycoprotein with a CAM motif. Cell 83, 433–442. doi: 10.1016/0092-8674(95)90121-3

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Isom, L. L., Scheuer, T., Brownstein, A. B., Ragsdale, D. S., Murphy, B. J., and Catterall, W. A. (1995b). Functional co-expression of the beta 1 and type IIA alpha subunits of sodium channels in a mammalian cell line. J. Biol. Chem. 270, 3306–3312. doi: 10.1074/jbc.270.7.3306

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kazen-Gillespie, K. A., Ragsdale, D. S., D'andrea, M. R., Mattei, L. N., Rogers, K. E., and Isom, L. L. (2000). Cloning, localization, and functional expression of sodium channel beta1A subunits. J. Biol. Chem. 275, 1079–1088. doi: 10.1074/jbc.275.2.1079

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Laedermann, C. J., Cachemaille, M., Kirschmann, G., Pertin, M., Gosselin, R. D., Chang, I., et al. (2013). Dysregulation of voltage-gated sodium channels by ubiquitin-ligase NEDD4-2 in neuropathic pain. J. Clin. Invest. 123, 3002–3013. doi: 10.1172/JCI68996

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lampert, A., O'reilly, A. O., Reeh, P., and Leffler, A. (2010). Sodium channelopathies and pain. Pflugers Arch. 460, 249–263. doi: 10.1007/s00424-009-0779-3

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Meadows, L. S., and Isom, L. L. (2005). Sodium channels as macromolecular complexes: implications for inherited arrhythmia syndromes. Cardiovasc. Res. 67, 448–458. doi: 10.1016/j.cardiores.2005.04.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Messner, D. J., and Catterall, W. A. (1985). The sodium channel from rat brain. separation and characterization of subunits. J. Biol. Chem. 260, 10597–10604.

Pubmed Abstract | Pubmed Full Text

Moremen, K. W., Tiemeyer, M., and Nairn, A. V. (2012). Vertebrate protein glycosylation: diversity, synthesis and function. Nat. Rev. Mol. Cell Biol. 13, 448–462. doi: 10.1038/nrm3383

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Morgan, K., Stevens, E. B., Shah, B., Cox, P. J., Dixon, A. K., Lee, K., et al. (2000). Beta 3: an additional auxiliary subunit of the voltage-sensitive sodium channel that modulates channel gating with distinct kinetics. Proc. Natl. Acad. Sci. U.S.A. 97, 2308–2313. doi: 10.1073/pnas.030362197

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Noda, M., Shimizu, S., Tanabe, T., Takai, T., Kayano, T., Ikeda, T., et al. (1984). Primary structure of Electrophorus electricus sodium channel deduced from cDNA sequence. Nature 312, 121–127. doi: 10.1038/312121a0

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Nuss, H. B., Chiamvimonvat, N., Perez-Garcia, M. T., Tomaselli, G. F., and Marban, E. (1995). Functional association of the beta 1 subunit with human cardiac (hH1) and rat skeletal muscle (mu 1) sodium channel alpha subunits expressed in Xenopus oocytes. J. Gen. Physiol. 106, 1171–1191. doi: 10.1085/jgp.106.6.1171

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Okuse, K., Malik-Hall, M., Baker, M. D., Poon, W. Y., Kong, H., Chao, M. V., et al. (2002). Annexin II light chain regulates sensory neuron-specific sodium channel expression. Nature 417, 653–656. doi: 10.1038/nature00781

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Parodi, A. J. (2000). Protein glucosylation and its role in protein folding. Annu. Rev. Biochem. 69, 69–93. doi: 10.1146/annurev.biochem.69.1.69

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Payandeh, J., Scheuer, T., Zheng, N., and Catterall, W. A. (2011). The crystal structure of a voltage-gated sodium channel. Nature 475, 353–358. doi: 10.1038/nature10238

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Recio-Pinto, E., Thornhill, W. B., Duch, D. S., Levinson, S. R., and Urban, B. W. (1990). Neuraminidase treatment modifies the function of electroplax sodium channels in planar lipid bilayers. Neuron 5, 675–684. doi: 10.1016/0896-6273(90)90221-Z

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ritchie, J. M., Black, J. A., Waxman, S. G., and Angelides, K. J. (1990). Sodium channels in the cytoplasm of Schwann cells. Proc. Natl. Acad. Sci. U.S.A. 87, 9290–9294. doi: 10.1073/pnas.87.23.9290

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sangameswaran, L., Fish, L. M., Koch, B. D., Rabert, D. K., Delgado, S. G., Ilnicka, M., et al. (1997). A novel tetrodotoxin-sensitive, voltage-gated sodium channel expressed in rat and human dorsal root ganglia. J. Biol. Chem. 272, 14805–14809. doi: 10.1074/jbc.272.23.14805

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Schmidt, J., Rossie, S., and Catterall, W. A. (1985). A large intracellular pool of inactive Na channel alpha subunits in developing rat brain. Proc. Natl. Acad. Sci. U.S.A. 82, 4847–4851. doi: 10.1073/pnas.82.14.4847

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Schmidt, J. W., and Catterall, W. A. (1987). Palmitylation, sulfation, and glycosylation of the alpha subunit of the sodium channel. Role of post-translational modifications in channel assembly. J. Biol. Chem. 262, 13713–13723.

Pubmed Abstract | Pubmed Full Text

Shah, B. S., Stevens, E. B., Gonzalez, M. I., Bramwell, S., Pinnock, R. D., Lee, K., et al. (2000). beta3, a novel auxiliary subunit for the voltage-gated sodium channel, is expressed preferentially in sensory neurons and is upregulated in the chronic constriction injury model of neuropathic pain. Eur. J. Neurosci. 12, 3985–3990. doi: 10.1046/j.1460-9568.2000.00294.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Smith, R. D., and Goldin, A. L. (1998). Functional analysis of the rat I sodium channel in xenopus oocytes. J. Neurosci. 18, 811–820.

Pubmed Abstract | Pubmed Full Text

Takahashi, N., Kikuchi, S., Dai, Y., Kobayashi, K., Fukuoka, T., and Noguchi, K. (2003). Expression of auxiliary beta subunits of sodium channels in primary afferent neurons and the effect of nerve injury. Neuroscience 121, 441–450. doi: 10.1016/S0306-4522(03)00432-9

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Tyrrell, L., Renganathan, M., Dib-Hajj, S. D., and Waxman, S. G. (2001). Glycosylation alters steady-state inactivation of sodium channel Nav1.9/NaN in dorsal root ganglion neurons and is developmentally regulated. J. Neurosci. 21, 9629–9637.

Pubmed Abstract | Pubmed Full Text

Valdivia, C. R., Medeiros-Domingo, A., Ye, B., Shen, W. K., Algiers, T. J., Ackerman, M. J., et al. (2010). Loss-of-function mutation of the SCN3B-encoded sodium channel citation{beta}3 subunit associated with a case of idiopathic ventricular fibrillation. Cardiovasc. Res. 86, 392–400. doi: 10.1093/cvr/cvp417

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Vijayaragavan, K., O'leary, M. E., and Chahine, M. (2001). Gating properties of Nav1.7 and Nav1.8 peripheral nerve sodium channels. J. Neurosci. 21, 7909–7918.

Pubmed Abstract | Pubmed Full Text

Vijayaragavan, K., Powell, A. J., Kinghorn, I. J., and Chahine, M. (2004). Role of auxiliary beta1-, beta2-, and beta3-subunits and their interaction with Na(v)1.8 voltage-gated sodium channel. Biochem. Biophys. Res. Commun. 319, 531–540. doi: 10.1016/j.bbrc.2004.05.026

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Waechter, C. J., Schmidt, J. W., and Catterall, W. A. (1983). Glycosylation is required for maintenance of functional sodium channels in neuroblastoma cells. J. Biol. Chem. 258, 5117–5123.

Pubmed Abstract | Pubmed Full Text

Yu, F. H., Westenbroek, R. E., Silos-Santiago, I., McCormick, K. A., Lawson, D., Ge, P., et al. (2003). Sodium channel β4, a new disulfide-linked auxiliary subunit with similarity to β2. J. Neurosci. 23, 7577–7585.

Pubmed Abstract | Pubmed Full Text

Zhang, Y., Hartmann, H. A., and Satin, J. (1999). Glycosylation influences voltage-dependent gating of cardiac and skeletal muscle sodium channels. J. Membr. Biol. 171, 195–207. doi: 10.1007/s002329900571

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Zhang, Z. N., Li, Q., Liu, C., Wang, H. B., Wang, Q., and Bao, L. (2008). The voltage-gated Na+ channel Nav1.8 contains an ER-retention/retrieval signal antagonized by the beta3 subunit. J. Cell Sci. 121, 3243–3252. doi: 10.1242/jcs.026856

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Zimmer, T., and Benndorf, K. (2002). The human heart and rat brain IIA Na+ channels interact with different molecular regions of the beta1 subunit. J. Gen. Physiol. 120, 887–895. doi: 10.1085/jgp.20028703

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Zimmer, T., Biskup, C., Bollensdorff, C., and Benndorf, K. (2002). The beta1 subunit but not the beta2 subunit colocalizes with the human heart Na+ channel (hH1) already within the endoplasmic reticulum. J. Membr. Biol. 186, 13–21. doi: 10.1007/s00232-001-0131-0

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Keywords: voltage-gated sodium channels (Navs), Navs β-subunits, glycosylation, biophysical properties, trafficking

Citation: Laedermann CJ, Syam N, Pertin M, Decosterd I and Abriel H (2013) β1- and β3- voltage-gated sodium channel subunits modulate cell surface expression and glycosylation of Nav1.7 in HEK293 cells. Front. Cell. Neurosci. 7:137. doi: 10.3389/fncel.2013.00137

Received: 24 May 2013; Paper pending published: 22 June 2013;
Accepted: 07 August 2013; Published online: 30 August 2013.

Edited by:

Dieter Wicher, Max Planck Institute for Chemical Ecology, Germany

Reviewed by:

Stefan H. Heinemann, Friedrich-Schiller-Universität, Germany
Todd Scheuer, University of Washington School of Medicine, USA

Copyright © 2013 Laedermann, Syam, Pertin, Decosterd and Abriel. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Hugues Abriel, Department of Clinical Research, University of Bern, Murtenstrasse 35, 3010 Bern, Switzerland e-mail: hugues.abriel@dkf.unibe.ch

These authors have contributed equally to this work and should be considered as co-first authors.

These authors have contributed equally to this work and should be considered as co-last authors.

Download