Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Enhanced Expression of WD Repeat-Containing Protein 35 via CaMKK/AMPK Activation in Bupivacaine-Treated Neuro2a Cells

  • Lei Huang ,

    honey326@qq.com

    Affiliations Department of Pharmacology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan, Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China

  • Fumio Kondo,

    Affiliation Department of Pharmacology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan

  • Masahiko Gosho,

    Affiliation Advanced Medical Research Center, Aichi Medical University, Nagakute, Aichi, Japan

  • Guo-Gang Feng,

    Affiliation Department of Pharmacology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan

  • Misako Harato,

    Affiliation Department of Anesthesiology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan

  • Zhong-yuan Xia,

    Affiliation Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China

  • Naohisa Ishikawa,

    Affiliation Department of Pharmacology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan

  • Yoshihiro Fujiwara,

    Affiliation Department of Anesthesiology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan

  • Shoshiro Okada

    Affiliation Department of Pharmacology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan

Expression of Concern

Following the publication of [1] concerns were raised in the following figure panels:

  • Fig 1B AMPKα panel appears similar to Fig 2A AMPKα panel
  • Fig 1B P-AMPKα panel contains vertical discontinuities across lanes
  • Fig 3A P-AMPKα panel contains vertical discontinuities across lanes
  • Fig 3A P-p38 panel contains a vertical discontinuity after lane 9
  • Fig 5A P-AMPKα panel contains a vertical discontinuity after lane 2
  • Fig 6B WDR35 panel contains a vertical discontinuity after lane 2

The authors explained that an incorrect image was used during preparation of Fig 1 and have provided a replacement image for the Fig 1B AMPKα panel. In the case of the Fig 1B P-AMPKα panel, the authors explained that samples were run in duplicate for Bupivacaine treatment and they removed the duplicate samples’ data in preparing the figure. However, the raw data provided by the authors suggests that the results across these duplicate samples were variable and those shown in the figure were not representative of the overall results.

The authors also noted that they spliced out lanes from the original image in preparing Fig 2A, and rearranged data in Fig 5A P-AMPKα panel for the purpose of presentation. For both of these panels, the authors provided the original blot data supporting the blots of concern. Individual-level data underlying all the graphs in the article are available.

However, authors have been unable to provide raw unadjusted blots underlying panels of concern in Figs 3A and 6B and are therefore not compliant with our data availability policy.

In light of the image concerns and the unavailability of data underlying the findings, the PLOS ONE Editors post this Expression of Concern.

22 Feb 2019: The PLOS ONE Editors (2019) Expression of Concern: Enhanced Expression of WD Repeat-Containing Protein 35 via CaMKK/AMPK Activation in Bupivacaine-Treated Neuro2a Cells. PLOS ONE 14(2): e0213041. https://doi.org/10.1371/journal.pone.0213041 View expression of concern

Abstract

We previously reported that bupivacaine induces reactive oxygen species (ROS) generation, p38 mitogen-activated protein kinase (MAPK) activation and nuclear factor-kappa B activation, resulting in an increase in expression of WD repeat-containing protein 35 (WDR35) in mouse neuroblastoma Neuro2a cells. However, the identity of signaling upstream of p38 MAPK pathways to WDR35 expression remains unclear. It has been shown that AMP-activated protein kinase (AMPK) can activate p38 MAPK through diverse mechanisms. In addition, several kinases acting upstream of AMPK have been identified including Ca2+/calmodulin-dependent protein kinase kinase (CaMKK). Recent studies reported that AMPK may be involved in bupivacaine-induced cytotoxicity in Schwann cells and in human neuroblastoma SH-SY5Y cells. The present study was undertaken to test whether CaMKK and AMPK are involved in bupivacaine-induced WDR35 expression in Neuro2a cells. Our results showed that bupivacaine induced activation of AMPK and p38 MAPK in Neuro2a cells. The AMPK inhibitors, compound C and iodotubercidin, attenuated the bupivacaine-induced activation of AMPK and p38 MAPK, resulting in an inhibition of the bupivacaine-induced increase in WDR35 expression. Treatment with the CaMKK inhibitor STO-609 also attenuated the bupivacaine-induced activation of AMPK and p38 MAPK, resulting in an inhibition of the bupivacaine-induced increase in WDR35 expression. These results suggest that bupivacaine activates AMPK and p38 MAPK via CaMKK in Neuro2a cells, and that the CaMKK/AMPK/p38 MAPK pathway is involved in regulating WDR35 expression.

Introduction

The family of WD repeat (WDR) proteins comprises a large number of proteins and is involved in a wide variety of cellular processes such as signal transduction, cell growth, proliferation, and apoptosis [1], [2]. WD repeat-containing protein 35 (WDR35) is a novel member of the WDR protein family [3]. Previously, we reported that enhanced WDR35 expression may mediate apoptosis in several animal models [4][6].

Bupivacaine-induced neurotoxicity has been associated with the generation of reactive oxygen species (ROS) [7] and activation of p38 mitogen-activated protein kinase (MAPK) [8], [9]. Recently, we demonstrated that bupivacaine induces ROS generation and p38 MAPK activation, resulting in an increase in WDR35 expression in mouse neuroblastoma Neuro2a cells [10]. More recently, we reported that bupivacaine induces the activation of nuclear factor-kappa B (NF-κB) in Neuro2a cells, and activation of NF-κB is involved in the bupivacaine-induced increase in WDR35 expression [11]. However, the identity of signaling upstream of p38 MAPK pathways to WDR35 expression remains unclear.

Many studies have demonstrated that AMP-activated protein kinase (AMPK) can activate p38 MAPK through diverse mechanisms [12][14]. AMPK is a heterotrimeric enzyme consisting of catalytic α- and regulatory β- and γ- subunits. Activation of AMPK requires phosphorylation of threonine (Thr172) in the activation loop of the α-subunit by upstream kinases [15][17]. AMPK is considered to be a regulator of cellular energy homeostasis, whereby it senses the metabolic status within a cell, especially under ATP deprivation, and is associated with the regulation of cellular stress in various cell types [17][19]. Recent studies demonstrated that AMPK may be involved in bupivacaine-induced cytotoxicity in Schwann cells [20] and in human neuroblastoma SH-SY5Y cells [21]. Several kinases acting upstream of AMPK have been identified including Ca2+/calmodulin-dependent protein kinase kinase (CaMKK), which can activate AMPK by phosphorylating the α-subunit at Thr172 [22], [23]. Recently, Pfisterer et al. [24] reported that CaMKK signaling via AMPK contributes to the regulation of WD-repeat protein interacting with phosphoinositides (WIPI)-1, another WDR protein family member, in starvation-induced autophagy. However, the involvement of CaMKK and AMPK to WDR35 expression has not been investigated. The present study was undertaken to test whether CaMKK and AMPK are involved in bupivacaine-induced WDR35 expression in Neuro2a cells. Our results suggest that AMPK is activated by bupivacaine in Neuro2a cells, and that the CaMKK/AMPK/p38 MAPK pathway is involved in regulating WDR35 expression.

Materials and Methods

Cell culture

Mouse neuroblastoma Neuro2a cells were purchased from the Health Science Research Resources Bank (Tokyo, Japan). The cells were maintained in RPMI-1640 medium (Sigma-Aldrich, St. Louis, MO, USA) containing 10% fetal bovine serum with 100 units/ml penicillin and 100 g/ml streptomycin (Gibco BRL, Grand Island, NY, USA). The cells were maintained at 37°C in a humidified atmosphere with 5% CO2. The culture medium was replaced every 2–3 days. To prepare cell suspensions, the cells were treated with trypsin (0.25%)-EDTA (1 mM) (Gibco BRL, Grand Island, NY, USA), transferred to a 6-cm culture plate at a density of 1.5×106 cells per dish, and cultured overnight.

In our previous study, we reported that bupivacaine dose-dependently increased WDR35 expression and that maximal WDR35 expression was observed with a concentration of 2 mM bupivacaine at 9 h [10]. As the maximal effect was reached at 9 h, the same 9 h time point of 2 mM bupivacaine treatment was used in the present study.

Measurement of ATP levels

Intracellular ATP levels were measured with a CellTiter-Glo Luminescent Cell Viability Assay kit (Promega, Madison, WI) according to the manufacturer's instructions. Briefly, after exposure of the cells to 2 mM bupivacaine for a period of 1 to 9 h, 100 µl of CellTiter-Glo reagent was added to 100 µl of the cell-containing medium. The contents were then mixed for 2 min to induce cell lysis, and the cells were incubated at room temperature for an additional 10 min to stabilize the luminescent signal. The luminance (RLU) was measured with a microplate reader (VersaMax, Molecular Devices, Sunnyvale, CA, USA).

Western blot analysis

Protein samples from cultured Neuro2a cells were homogenized in sample buffer [50 mM Tris-HCl (pH 6.8), 0.2 M DTT, 2% sodium dodecyl sulfate (SDS), 10% glycerol, 0.1% bromophenol blue (BPB)] containing a mixture of protease inhibitors (Complete Protease Inhibitor Cocktail, Roche Applied Sciences, Mannheim, Germany) and heated in boiling water for 5 min. Proteins were separated by SDS-PAGE and transferred to PVDF membranes (Immobilon-P, Millipore, Bedford, MA, USA). The membranes were blocked in 5% w/v bovine serum albumin in Tris-buffered saline (TBST, 0.05 M Tris, 0.15 M NaCl, 0.1% Tween-20, pH 7.4) overnight at 4°C. These membranes were probed with an anti-WDR35 peptide antibody (amino acids 459-473, 1∶500), which was designed, produced, and purified by Medical & Biological Laboratories (Nagoya, Japan), or with antibodies against AMPKα, phospho-AMPKα (Thr172) (P-AMPKα), p38 MAPK, phospho-p38 (P-p38) MAPK, or β-actin (Cell Signaling Technology, Danvers, MA, USA; 1∶1000). Detection was performed with the Western blotting reagent ECL Prime (GE Healthcare, Buckinghamshire, UK). Protein levels were quantified by densitometric scanning with the Gel-Pro Analyzer (Media Cybernetics, Inc., USA) and expressed as the ratio to β-actin levels as described previously [10], [11].

Quantitative real-time polymerase chain reaction (qPCR) analysis

Total RNA (1 µg) was extracted from cultured Neuro2a cells with TRIzol reagent (Invitrogen, Carlsbad, CA, USA) and reverse transcribed with the ReverTra Ace qPCR RT kit (Toyobo, Osaka, Japan). qPCR was performed with the ABI StepOne Plus real-time PCR system and a TaqMan Gene Expression Assay (Applied Biosystems, Tokyo, Japan) according to the manufacturer's instructions. The primers and TaqMan MGB probe for mouse WDR35 (Mm00552650_m1) were purchased from Applied Biosystems. The amount of WDR35 PCR product was calculated relative to the internal controls β-actin (ACTB, Mm00607939_s1; Applied Biosystems), glyceraldehyde-3-phosphate dehydrogenase (GAPDH, Mm99999915_g1), or 18S ribosomal RNA (18S rRNA, Mm03928990_g1), and was compared between experimental and control groups by the ΔΔCT method, as reported previously [10], [11]. The difference in internal controls did not affect the results in the qPCR experiments.

Statistical analysis

All results are expressed as the mean ± standard error of the mean (SEM). Data were analyzed by one-way or two-way analysis of variance (ANOVA) followed by Tukey's post-hoc test. Statistical analysis for time effect was performed by repeated measures one-way ANOVA. Treatment and time effects were analyzed using repeated measures two-way ANOVA including treatment group and time period as factors and the interaction term between treatment group and time period. Differences were considered significant at P<0.05. All statistical analyses were done with the SAS 9.3 (SAS institute, Cary, USA).

Results

Bupivacaine induces AMPK activation in Neuro2a cells

We first measured the intracellular ATP levels in bupivacaine-treated Neuro2a cells. As shown in Figure 1A, treatment with 2 mM bupivacaine significantly reduced intracellular ATP levels in Neuro2a cells in a time-dependent manner (P<0.001 at 3 h and later; Figure 1A).

thumbnail
Figure 1. Effect of bupivacaine on intracellular ATP levels and AMPK activation.

Neuro2a cells were treated with 2(A) Intracellular levels of ATP were measured with a luminescence assay (n = 4 per group). (B) Expression of phospho-AMPKα (P-AMPKα) and AMPK following cell exposure to bupivacaine was measured by Western blotting (n = 3 per group). ***P<0.001 vs. control (not treated with bupivacaine). All post hoc pairwise comparisons were performed by Tukey's test with repeated measures one-way ANOVA.

https://doi.org/10.1371/journal.pone.0098185.g001

To determine whether AMPK is activated in bupivacaine-treated Neuro2a cells, we then examined AMPKα and phospho-AMPKα (P-AMPKα) protein expression in bupivacaine-treated Neuro2a cells by Western blotting and expressed the results as the P-AMPKα/AMPKα ratio. As shown in Figure 1B, treatment of Neuro2a cells with 2 mM bupivacaine significantly increased phosphorylation levels of AMPKα at 1 to 9 h (P<0.001 at 1 h and later).

AMPK inhibitors, compound C and iodotubercidin, attenuate the bupivacaine-induced increase in AMPK and p38 MAPK activity in Neuro2a cells

In order to explore the relationship between AMPK and p38 MAPK activation in bupivacaine-treated Neuro2a cells, cells were treated for 1 h with the AMPK inhibitor compound C (10 µM; Calbiochem, La Jolla, CA, USA) or iodotubercidin (1 µM; Abcam, Cambridge, MA, USA), followed by bupivacaine (2 mM) for 1 to 9 h. As shown in Figure 2A and 2B, compound C significantly attenuated the bupivacaine-induced increase in phosphorylation levels of AMPKα at time points from 3 to 9 h (P<0.05 and P<0.001). Compound C also significantly attenuated the bupivacaine-induced increase in phosphorylation levels of p38 MAPK at time points from 3 to 9 h (P<0.05 and P<0.01; Figure 2A and 2C).

thumbnail
Figure 2. Effect of compound C on bupivacaine-induced AMPK and p38 MAPK activity.

Neuro2a cells were treated with AMPK inhibitor compound C (10 µM) for 1 h, followed by bupivacaine (2 mM) for 9 h. (A) Expression of phospho-AMPKα (P-AMPKα), AMPK, phospho-p38 (P-p38) and p38 was measured by Western blotting (n = 3 per group). The bar diagram shows the ratio of phospho-AMPKα to AMPK (B) and the ratio of P-p38 to p38 (C). *P<0.05, **P<0.01 and ***P<0.001 vs. control (not treated with bupivacaine), #P<0.05, ##P<0.01 and ###P<0.001 vs. absence of compound C. All post hoc comparisons were performed by Tukey's test with repeated measures two-way ANOVA including compound C group and time period as factors and the interaction term between the two factors.

https://doi.org/10.1371/journal.pone.0098185.g002

Treatment with iodotubercidin significantly attenuated the bupivacaine-induced increase in phosphorylation levels of AMPKα at time points from 1 to 9 h (P<0.05, P<0.01 and P<0.001; Figure 3A and 3B). Iodotubercidin also significantly attenuated the bupivacaine-induced increase in phosphorylation levels of p38 MAPK at time points from 3 to 9 h (P<0.01 and P<0.001; Figure 3A and 3C).

thumbnail
Figure 3. Effect of iodotubercidin on bupivacaine-induced AMPK and p38 MAPK activity.

Neuro2a cells were treated with AMPK inhibitor iodotubercidin (1 µM) for 1 h, followed by bupivacaine (2 mM) for 9 h. (A) Expression of phospho-AMPKα (P-AMPKα), AMPK, phospho-p38 (P-p38) and p38 was measured by Western blotting (n = 3 per group). The bar diagram shows the ratio of phospho-AMPKα to AMPK (B) and the ratio of P-p38 to p38 (C). *P<0.05, **P<0.01 and ***P<0.001 vs. control (not treated with bupivacaine), #P<0.05, ##P<0.01 and ###P<0.001 vs. absence of iodotubercidin. All post hoc comparisons were performed by Tukey's test with repeated measures two-way ANOVA including iodotubercidin group and time period as factors and the interaction term between the two factors.

https://doi.org/10.1371/journal.pone.0098185.g003

AMPK inhibitors, compound C and iodotubercidin, attenuate the bupivacaine-induced increase in WDR35 expression in Neuro2a cells

In order to explore the relationship between WDR35 expression and AMPK activation, Neuro2a cells were treated for 1 h with compound C (5, 10 and 20 µM) or iodotubercidin (0.5, 1 and 10 µM), followed by bupivacaine (2 mM) for 9 h. As shown in Figure 4A, compound C significantly attenuated the bupivacaine-induced increase in WDR35 mRNA expression in a dose-dependent manner (P<0.001). Treatment with 10 µM compound C significantly attenuated the bupivacaine-induced increase in WDR35 protein expression (P<0.05; Figure 4B).

thumbnail
Figure 4. Effect of compound C and iodotubercidin on bupivacaine-induced WDR35 expression.

(A) Neuro2a cells were treated with various concentrations of compound C for 1 h, followed by bupivacaine (2 mM) for 9 h. WDR35 mRNA expression was analyzed by qPCR and expressed relative to the level of β-actin mRNA (n = 4 per group). All post hoc comparisons were performed by Tukey's test with one-way ANOVA. (B) WDR35 protein expression with or without 10 µM compound C was analyzed by Western blotting (n = 4 per group). All post hoc comparisons were performed by Tukey's test with two-way ANOVA including bupivacaine and compound C groups as factors and the interaction term between the two factors. (C) Cells were treated with various concentrations of iodotubercidin for 1 h, followed by bupivacaine (2 mM) for 9 h. WDR35 mRNA expression was analyzed by qPCR (n = 4 per group). All post hoc comparisons were performed by Tukey's test with one-way ANOVA. (D) WDR35 protein expression with or without 1 µM iodotubercidin was analyzed by Western blotting (n = 3 per group). All post hoc comparisons were performed by Tukey's test with two-way ANOVA including bupivacaine and iodotubercidin groups as factors and the interaction term between the two factors. **P<0.01 and ***P<0.001 vs. control (not treated with bupivacaine), #P<0.05 and ###P<0.001 vs. treatment with bupivacaine alone.

https://doi.org/10.1371/journal.pone.0098185.g004

Treatment with iodotubercidin significantly attenuated the bupivacaine-induced increase in WDR35 mRNA expression in a dose-dependent manner (P<0.001; Figure 4C). Iodotubercidin also significantly attenuated the bupivacaine-induced increase in WDR35 protein expression (P<0.05; Figure 4D).

STO-609, a CaMKK inhibitor, attenuates the bupivacaine-induced increase in AMPK and p38 MAPK activity in Neuro2a cells

Several kinases acting upstream of AMPK have been identified including CaMKK which is highly expressed in neurons [25][27]. In order to examine whether CaMKK is involved in bupivacaine-induced AMPK activation, we further investigated the effects of STO-609, a CaMKK inhibitor, on AMPKα protein expression in bupivacaine-treated Neuro2a cells. Neuro2a cells were treated with STO-609 (50 µM; Calbiochem, La Jolla, CA, USA) for 1 h, followed by bupivacaine (2 mM) for 1 to 9 h. As shown in Figure 5A and 5B, treatment with STO-609 significantly attenuated the bupivacaine-induced increase in phosphorylation levels of AMPKα at time points from 1 to 9 h (P<0.01 and P<0.001). Similarly, STO-609 significantly attenuated the bupivacaine-induced increase in phosphorylation levels of p38 MAPK at time points from 1 to 9 h (P<0.01 and P<0.001; Figure 5A and 5C).

thumbnail
Figure 5. Effect of STO-609 on bupivacaine-induced AMPK and p38 MAPK activity.

Neuro2a cells were treated with CaMKK inhibitor STO-609 (50 µM) for 1 h, followed by bupivacaine (2 mM) for 9 h. (A) Expression of phospho-AMPKα (P-AMPKα), AMPK, phospho-p38 (P-p38) and p38 was measured by Western blotting (n = 3 per group). The bar diagram shows the ratio of phospho-AMPKα to AMPK (B) and the ratio of P-p38 to p38 (C). **P<0.01 and ***P<0.001 vs. control (not treated with bupivacaine), ##P<0.01 and ###P<0.001 vs. absence of STO-609. All post hoc comparisons were performed by Tukey's test with repeated measures two-way ANOVA including STO-609 group and time period as factors and the interaction term between the two factors.

https://doi.org/10.1371/journal.pone.0098185.g005

STO-609 attenuates the bupivacaine-induced increase in WDR35 expression in Neuro2a cells

In order to explore the relationship between WDR35 expression and CaMKK, Neuro2a cells were treated for 1 h with STO-609 (5, 20 and 50 µM), followed by bupivacaine (2 mM) for 9 h. As shown in Figure 6A, STO-609 significantly attenuated the bupivacaine-induced increase in WDR35 mRNA expression in a dose-dependent manner (P<0.001). In addition, treatment with 50 µM STO-609 significantly attenuated the bupivacaine-induced increase in WDR35 protein expression (P<0.01; Figure 6B).

thumbnail
Figure 6. Effect of STO-609 on bupivacaine-induced WDR35 expression.

(A) Neuro2a cells were treated with various concentrations of STO-609 for 1 h, followed by bupivacaine (2 mM) for 9 h. WDR35 mRNA expression was analyzed by qPCR and expressed relative to the level of β-actin mRNA (n = 4 per group). All post hoc comparisons were performed by Tukey's test with one-way ANOVA. (B) WDR35 protein expression with or without 50 µM STO-609 was analyzed by Western blotting (n = 3 per group). All post hoc comparisons were performed by Tukey's test with two-way ANOVA including bupivacaine and STO-609 groups as factors and the interaction term between the two factors. **P<0.01 and ***P<0.001 vs. control (not treated with bupivacaine), ##P<0.01 and ###P<0.001 vs. absence of STO-609.

https://doi.org/10.1371/journal.pone.0098185.g006

Discussion

We previously reported that bupivacaine induces ROS generation, p38 MAPK activation and NF-κB activation, resulting in an increase in expression of WDR35 in Neuro2a cells [10], [11]. In the present study, we investigated the relevance of kinases acting upstream of p38 MAPK to WDR35 expression. Bupivacaine induced activation of AMPK and p38 MAPK in Neuro2a cells. The AMPK inhibitors, compound C and iodotubercidin, attenuated the bupivacaine-induced activation of AMPK and p38 MAPK, resulting in an inhibition of the bupivacaine-induced increase in WDR35 expression. Treatment with the CaMKK inhibitor STO-609 also attenuated the bupivacaine-induced activation of AMPK and p38 MAPK, resulting in an inhibition of the bupivacaine-induced increase in WDR35 expression. Collectively, these results suggest that the CaMKK/AMPK/p38 MAPK pathway is involved in regulating WDR35 expression in bupivacaine-treated Neuro2a cells.

At the molecular level, AMPK is considered to regulate energy balance in response to decreases in ATP, which is important for regulating cellular homeostasis under various kinds of stress [17], [22], [23]. In the present study, we showed that 2 mM bupivacaine reduced intracellular ATP levels in a time-dependent manner in Neuro2a cells. Our results are consistent with a previous report that showed the same dose of bupivacaine decreased intracellular ATP levels in B16 melanoma cells [28]. It has been reported that bupivacaine increased AMPK activity in Schwann cells [20], and we subsequently confirmed the activation of AMPK in bupivacaine-treated Neuro2a cells. These results demonstrated that bupivacaine reduced intracellular ATP levels and increased AMPK activity in Neuro2a cells.

In the present study, we showed that the AMPK inhibitors, compound C and iodotubercidin, attenuated the bupivacaine-induced increase in AMPK and p38 MAPK activity in Neuro2a cells. These results are in agreement with previous reports that AMPK activation could lead to p38 MAPK activation in airway epithelial cells [12], osteoblasts [13] and muscle cells [14]. CaMKK, one of the upstream AMPK kinases, is abundantly expressed in neurons and is activated by an increase in intracellular Ca2+ concentration [25][27]. In neurons, Ca2+ balance dysfunction has been shown to trigger AMPK signaling [23]. Recently, Wen et al. [29] reported that bupivacaine treatment leads to an increase in intracellular Ca2+ concentration in SH-SY5Y cells. We thus examined the effects of STO-609, a CaMKK inhibitor, on AMPK and p38 MAPK activity. STO-609 treatment attenuated the bupivacaine-induced increase in AMPK and p38 MAPK activity. Collectively, these results indicate that CaMKK is involved in bupivacaine-induced AMPK and p38 MAPK activation.

Recently, we demonstrated that bupivacaine induces ROS generation and p38 MAPK activation, resulting in an increase in WDR35 expression in Neuro2a cells [10]. To determine whether AMPK activation is involved in bupivacaine-induced WDR35 expression, we examined the effects of the AMPK inhibitors, compound C and iodotubercidin, on WDR35 expression in bupivacaine-treated Neuro2a cells. These inhibitors significantly attenuated the bupivacaine-induced increase in WDR35 expression in Neuro2a cells. Moreover, a separate research group reported that CaMKK signaling via AMPK contributes to the regulation of another WDR protein, WIPI-1 [24]. Here, we showed that the CaMKK inhibitor, STO-609, attenuated the bupivacaine-induced increase in WDR35 expression in Neuro2a cells. Collectively, these results indicate that the CaMKK/AMPK/p38 MAPK pathway is involved in regulating WDR35 expression in bupivacaine-treated Neuro2a cells.

The STO-609 treatment only partially altered the transcriptional activation of WDR35 while potently inhibited AMPK and p38 MAPK, a downstream target of CaMKK. This result is surprising since acting more downstream in the cascade is supposed to elicit less dramatic effects on the activation of transcriptional programs. Although compound C and STO-609 have been shown to have some “off target” effects [30], these findings raise an interesting possibility that another signaling pathway might be involved in the induction of WDR35 in response to bupivacaine.

Recent studies have demonstrated that the WDR35 gene is involved in several human diseases such as type 2 diabetes [31], acute lymphoblastic leukemia [32], coronary artery disease [33], and Sensenbrenner syndrome [34]. However, the regulation of WDR35 expression by intracellular signaling pathways has not been fully elucidated. In the present study, we provide the first evidence of the sequence of events in bupivacaine-induced WDR35 expression by in vitro biochemical and pharmacological examinations, and show that CaMKK/AMPK signaling acts upstream to regulate WDR35 expression. Since the function of WDR35 is still unknown, further studies incorporating (1) proteome analysis of proteins interacting with WDR35 and (2) promoter analysis of the WDR35 gene will help to understand the role of WDR35.

In conclusion, our results indicate that bupivacaine activates AMPK and p38 MAPK via CaMKK in Neuro2a cells. Taken together with our previous study, the CaMKK/AMPK/p38 MAPK/NF-κB pathway is involved in regulating WDR35 expression in bupivacaine-treated Neuro2a cells (Figure 7).

thumbnail
Figure 7. Schematic diagram illustrating the proposed signaling pathway involved in bupivacaine-induced WDR35 expression in Neuro2a cells.

https://doi.org/10.1371/journal.pone.0098185.g007

Author Contributions

Conceived and designed the experiments: LH FK SO. Performed the experiments: LH GF MH. Analyzed the data: LH MG ZX NI YF. Contributed to the writing of the manuscript: LH FK MG SO.

References

  1. 1. Neer EJ, Schmidt CJ, Nambudripad R, Smith TF (1994) The ancient regulatory-protein family of WD-repeat proteins. Nature 371: 297–300.
  2. 2. Smith TF, Gaitatzes C, Saxena K, Neer EJ (1999) The WD repeat: a common architecture for diverse functions. Trends Biochem Sci 24: 181–185.
  3. 3. Feng GG, Li C, Huang L, Tsunekawa K, Sato Y, et al. (2010) Naofen, a novel WD40-repeat protein, mediates spontaneous and tumor necrosis factor-induced apoptosis. Biochem Biophys Res Commun 394: 153–157.
  4. 4. Sato Y, Feng GG, Huang L, Fan JH, Li C, et al. (2010) Enhanced expression of naofen in kidney of streptozotocin-induced diabetic rats: possible correlation to apoptosis of tubular epithelial cells. Clin Exp Nephrol 14: 205–212.
  5. 5. Fan JH, Feng GG, Huang L, Tsunekawa K, Honda T, et al. (2012) Role of naofen in apoptosis of hepatocytes induced by lipopolysaccharide through mitochondrial signaling in rats. Hepatol Res 42: 696–705.
  6. 6. Tsunekawa K, Kondo F, Okada T, Feng GG, Huang L, et al. (2013) Enhanced expression of WD repeat-containing protein 35 (WDR35) stimulated by domoic acid in rat hippocampus: involvement of reactive oxygen species generation and p38 mitogen-activated protein kinase activation. BMC Neurosci 14: 4.
  7. 7. Park CJ, Park SA, Yoon TG, Lee SJ, Yum KW, et al. (2005) Bupivacaine induces apoptosis via ROS in the Schwann cell line. J Dent Res 84: 852–857.
  8. 8. Lirk P, Haller I, Colvin HP, Lang L, Tomaselli B, et al. (2008) In vitro, inhibition of mitogen-activated protein kinase pathways protects against bupivacaine- and ropivacaine-induced neurotoxicity. Anesth Analg 106: 1456–1464.
  9. 9. Lu J, Xu SY, Zhang QG, Xu R, Lei HY (2011) Bupivacaine induces apoptosis via mitochondria and p38 MAPK dependent pathways. Eur J Pharmacol 657: 51–58.
  10. 10. Harato M, Huang L, Kondo F, Tsunekawa K, Feng GG, et al. (2012) Bupivacaine-induced apoptosis independently of WDR35 expression in mouse neuroblastoma Neuro2a cells. BMC Neurosci 13: 149.
  11. 11. Huang L, Kondo F, Harato M, Feng GG, Ishikawa N, et al. (2014) Enhanced expression of WD repeat-containing protein 35 via nuclear factor-kappa B activation in bupivacaine-treated Neuro2a cells. PLoS One 9: e86336.
  12. 12. Bae CH, Kim JW, Ye SB, Song SY, Kim YW, et al. (2011) AMPK induces MUC5B expression via p38 MAPK in NCI-H292 airway epithelial cells. Biochem Biophys Res Commun 409: 669–674.
  13. 13. Kondo A, Otsuka T, Kato K, Natsume H, Kuroyanagi G, et al. (2012) AMP-activated protein kinase inhibitor decreases prostaglandin F-stimulated interleukin-6 synthesis through p38 MAP kinase in osteoblasts. Int J Mol Med 30: 1487–1492.
  14. 14. Kim SH, Hwang JT, Park HS, Kwon DY, Kim MS (2013) Capsaicin stimulates glucose uptake in C2C12 muscle cells via the reactive oxygen species (ROS)/AMPK/p38 MAPK pathway. Biochem Biophys Res Commun 439: 66–70.
  15. 15. Carling D, Sanders MJ, Woods A (2008) The regulation of AMP-activated protein kinase by upstream kinases. Int J Obes 32: S55–S59.
  16. 16. Amato S, Man HY (2011) Bioenergy sensing in the brain: the role of AMP-activated protein kinase in neuronal metabolism, development and neurological diseases. Cell Cycle 10: 3452–3460.
  17. 17. Cai Z, Yan LJ, Li K, Quazi SH, Zhao B (2012) Roles of AMP-activated protein kinase in Alzheimer's disease. Neuromolecular Med 14: 1–14.
  18. 18. Weisová P, Dávila D, Tuffy LP, Ward MW, Concannon CG, et al. (2011) Role of 5'-adenosine monophosphate-activated protein kinase in cell survival and death responses in neurons. Antioxid Redox Signal 14: 1863–1876.
  19. 19. Carling D, Thornton C, Woods A, Sanders MJ (2012) AMP-activated protein kinase: new regulation, new roles? Biochem J 445: 11–27.
  20. 20. Lee SJ, Shin TJ, Kang IS, Ha JH, Lee SC, et al. (2010) AMPK attenuates bupivacaine-induced neurotoxicity. J Dent Res 89: 797–801.
  21. 21. Lu J, Xu SY, Zhang QG, Lei HY (2011) Bupivacaine induces reactive oxygen species production via activation of the AMP-activated protein kinase-dependent pathway. Pharmacology 87: 121–129.
  22. 22. Gruzman A, Babai G, Sasson S (2009) Adenosine Monophosphate-Activated Protein Kinase (AMPK) as a New Target for Antidiabetic Drugs: A Review on Metabolic, Pharmacological and Chemical Considerations. Rev Diabet Stud 6: 13–36.
  23. 23. Salminen A, Kaarniranta K, Haapasalo A, Soininen H, Hiltunen M (2011) AMP-activated protein kinase: a potential player in Alzheimer's disease. J Neurochem 118: 460–474.
  24. 24. Pfisterer SG, Mauthe M, Codogno P, Proikas-Cezanne T (2011) Ca2+/calmodulin-dependent kinase (CaMK) signaling via CaMKI and AMP-activated protein kinase contributes to the regulation of WIPI-1 at the onset of autophagy. Mol Pharmacol 80: 1066–1075.
  25. 25. Nakamura Y, Okuno S, Kitani T, Otake K, Sato F, et al. (2001) Immunohistochemical localization of Ca2+/calmodulin-dependent protein kinase kinase beta in the rat central nervous system. Neurosci Res 39: 175–188.
  26. 26. Vinet J, Carra S, Blom JM, Harvey M, Brunello N, et al. (2003) Cloning of mouse Ca2+/calmodulin-dependent protein kinase kinase beta (CaMKKbeta) and characterization of CaMKKbeta and CaMKKalpha distribution in the adult mouse brain. Brain Res Mol Brain Res 111: 216–221.
  27. 27. Racioppi L, Means AR (2012) Calcium/calmodulin-dependent protein kinase kinase 2: roles in signaling and pathophysiology. J Biol Chem 287: 31658–31665.
  28. 28. Karniel M, Beitner R (2000) Local anesthetics induce a decrease in the levels of glucose 1, 6-bisphosphate, fructose 1,6-bisphosphate, and ATP, and in the viability of melanoma cells. Mol Genet Metab 69: 40–45.
  29. 29. Wen X, Xu S, Liu H, Zhang Q, Liang H, et al. (2013) Neurotoxicity induced by bupivacaine via T-type calcium channels in SH-SY5Y cells. PLoS One 8: e62942.
  30. 30. Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, et al. (2007) The selectivity of protein kinase inhibitors: a further update. Biochem J 408: 297–315.
  31. 31. Berisha SZ, Serre D, Schauer P, Kashyap SR, Smith JD (2011) Changes in whole blood gene expression in obese subjects with type 2 diabetes following bariatric surgery: a pilot study. PLoS One 6: e16729.
  32. 32. Nordlund J, Milani L, Lundmark A, Lönnerholm G, Syvänen AC (2012) DNA methylation analysis of bone marrow cells at diagnosis of acute lymphoblastic leukemia and at remission. PLoS One 7: e34513.
  33. 33. Lu X, Wang L, Chen S, He L, Yang X, et al. (2012) Genome-wide association study in Han Chinese identifies four new susceptibility loci for coronary artery disease. Nat Genet 44: 890–894.
  34. 34. Bacino CA, Dhar SU, Brunetti-Pierri N, Lee B, Bonnen PE (2012) WDR35 mutation in siblings with Sensenbrenner syndrome: a ciliopathy with variable phenotype. Am J Med Genet A 158A: 2917–2924.