Background:

The histone deacetylases (HDACs) have been implicated in pain hypersensitivity. This study investigated the potential involvement of an HDAC4-related mechanism in the spinal nerve ligation (SNL)-induced nociceptive hypersensitivity.

Methods:

The left L5 to L6 spinal nerves of 627 adult male Sprague–Dawley rats were surgically ligated. The withdrawal threshold of hind paws and the abundances, cellular location, and interactions of proteins in the dorsal horn were assayed before and after surgery. The 14-3-3β-targeting small-interfering RNA, a serum- and glucocorticoid-inducible kinase 1 (SGK1) antagonist, or an HDAC inhibitor was spinally injected to elucidate the role of 14-3-3β, SGK1, and HDAC4.

Results:

Without affecting the HDAC4 level, SNL provoked SGK1 phosphorylation (mean ± SEM from 0.24 ± 0.02 to 0.78 ± 0.06 at day 7, n = 6), HDAC4 phosphorylation (from 0.38 ± 0.03 to 0.72 ± 0.06 at day 7, n = 6), 14-3-3β expression (from 0.53 ± 0.09 to 0.88 ± 0.09 at day 7, n = 6), cytoplasmic HDAC4 retention (from 1.18 ± 0.16 to 1.92 ± 0.11 at day 7, n = 6), and HDAC4-14-3-3β coupling (approximately 2.4-fold) in the ipsilateral dorsal horn in association with behavioral allodynia. Knockdown of spinal 14-3-3β expression prevented the SNL-provoked HDAC4 retention (from 1.89 ± 0.15 to 1.32 ± 0.08 at day 7, n = 6), HDAC4-14-3-3β coupling (approximately 0.6-fold above SNL 7D), and behavioral allodynia (from 0.16 ± 0.3 to 6 ± 1.78 at day 7, n = 7), but not SGK1 (from 0.78 ± 0.06 to 0.71 ± 0.04 at day 7, n = 6) or HDAC4 (from 0.75 ± 0.15 to 0.68 ± 0.11 at day 7, n = 6) phosphorylation.

Conclusion:

Neuropathic pain maintenance involves the spinal SGK1 activation–dependent HDAC4 phosphorylation and its subsequent association with 14-3-3β that promotes cytoplasmic HDAC4 retention in dorsal horn neurons.

In a rat model of neuropathic pain, histone deacetylases 4 phosphorylation led to its cytoplasmic retention due to phosphorylation-dependent interaction with 14-3-3β. Inhibition of histone deacetylases phosphorylation reduced allodynia and prevented its cytoplasmic translocation, suggesting a novel therapeutic target for neuropathic pain.

What We Already Know about This Topic
  • Pain-associated hypersensitivity (allodynia) involves activity-dependent effects on spinal neuroplasticity

  • Regulation of histone acetylation by histone deacetylase (HDAC) regulates gene transcription contribute to neuropathic pain

What This Article Tells Us That Is New
  • In a rat model of neuropathic pain, HDAC4 phosphorylation led to its cytoplasmic retention due to phosphorylation- dependent interaction with 14-3-3β

  • Inhibition of histone deacetylase phosphorylation reduced allodynia and prevented its cytoplasmic translocation, suggesting a novel therapeutic target for neuropathic pain

ACCUMULATING evidence has revealed that activity-dependent neural plasticity in the spinal cord underlies pain hypersensitivity1 ; therefore, both clinicians and scientists are highly interested in developing drugs targeted to the pain-associated plasticity to provide medical therapeutics for pain relief.

Intrathecal administration with a class IIa histone deacetylase (HDAC) inhibitor attenuated complete Freund adjuvant–induced inflammation hyperalgesia2  and deletion of the functional domain of HDAC4, a member of the class IIa HDACs,3  and increased the response latency to nociceptive stimuli,4  indicating HDAC4 is a crucial contributor to the spinal machinery underlying heightening of nociception.

Unlike other HDACs that either reside in the nucleus or are mostly in the cytoplasm, class IIa HDACs actively shuttle between the nucleus and cytoplasm.5  The nucleocytoplasmic shuttling of HDAC4 is primarily regulated through the phosphorylation of its serine residues, that is, phosphorylation prompts HDAC4 export from the nucleus that subsequently recruits 14-3-3 to anchor the resulting complex and thereby restricts HDAC4 location to the cytoplasm.6  Conversely, dephosphorylation uncouples HDAC4 from 14-3-3, thereby allowing HDAC4 to shuttle into the nucleus.7  By binding to the phospho-serine-/threonine-containing motifs of a variety of proteins, 14-3-3, a ubiquitous phosphor-binding protein,8  regulates a wide range of cellular processes including intracellular protein trafficking.9  Neuropathic injury selectively up-regulated 14-3-3 expression in rat dorsal horn,10  and 14-3-3β-HDAC4 interactions subsequent to HDAC4 phosphorylation have been linked to diverse physiological/pathological cellular programs.11  Nevertheless, it remains unclear that whether spinal HDAC4 phosphorylation and the following HDAC4-14-3-3β coupling could participate in the development of nociception hypersensitivity via regulating the subcellular HDAC4 distribution.

By phosphorylating the serine residue of neural precursor cell-expressed developmentally down-regulated protein 4-2 (Nedd4-2), serum- and glucocorticoid-inducible kinase 1 (SGK1), a serine/threonine kinase,12  facilitates the association of Nedd4-2 with 14-3-3.9  Interestingly, our studies have previously demonstrated that spinal SGK1 crucially participates in the nociception-associated plasticity by regulating serine/threonine phosphorylation–dependent protein trafficking13  and anchoring.14  Together, these several observations prompted us to test whether SGK1-dependent HDAC4 phosphorylation, coupling with 14-3-3β, and cytosolic accumulation in dorsal horn neurons contribute to the neuropathic injury–induced nociceptive hypersensitivity (fig. 1).

Fig. 1.

Schematic diagram showing serum- and glucocorticoid-inducible kinase 1 (SGK1), histone deacetylase (HDAC4), and 14-3-3β contribute to the neuropathic injury–associated nociceptive hypersensitivity. Possibly through regulating neurotransmitter release, spinal nerve ligation (SNL) activates SGK1 that provokes HDAC4 phosphorylation and the subsequent HDAC4-14-3-3β coupling, which restricts HDAC4 location to the cytoplasm of dorsal horn neurons. Reduced inhibition of HDAC4 on DNA transcription might up-regulate plasticity-associated protein, such as the N-methyl-d-aspartate receptor (NMDAR).

Fig. 1.

Schematic diagram showing serum- and glucocorticoid-inducible kinase 1 (SGK1), histone deacetylase (HDAC4), and 14-3-3β contribute to the neuropathic injury–associated nociceptive hypersensitivity. Possibly through regulating neurotransmitter release, spinal nerve ligation (SNL) activates SGK1 that provokes HDAC4 phosphorylation and the subsequent HDAC4-14-3-3β coupling, which restricts HDAC4 location to the cytoplasm of dorsal horn neurons. Reduced inhibition of HDAC4 on DNA transcription might up-regulate plasticity-associated protein, such as the N-methyl-d-aspartate receptor (NMDAR).

Close modal

Animal Preparations

The animal procedures in this study were conducted in accordance with the guidelines of the International Association for the Study of Pain15  and were reviewed and approved by the Institutional Review Board of National Chung-Hsing University, Taichung, Taiwan, Republic of China. A total of 627 adult male Sprague–Dawley rats (180 to 230 g body weight) were used throughout this study. Animal allocation to treatment groups was randomized by a computer with the use of research randomizer, and the sample size of each group was based on our previous experience. There were 119, 294, 14, and 192 rats, respectively, for behavioral test, Western blot, immunohistochemistry, and coprecipitation analyses performed by investigators blinded to treatment groups. There were eight animals that showed neurological deficits after implantation of a catheter and were excluded for statistical analysis. Thus, there were a total of 619 animals used for statistical analysis.

Spinal Nerve Ligation

After anesthesia (isoflurane, induction 5%, maintenance 2%), the left L5 to L6 spinal nerves were carefully isolated and was tightly ligated with 6-0 silk sutures 2 to 5 mm distal to the dorsal root ganglia.13,14,16  The procedures for the sham operation were identical to the nerve ligation group, excepting the silk sutures were left unligated.

Intrathecal Catheter

An PE-10 silastic tubing was implanted to the lumbar enlargement of the spinal cord. Animals were allowed to recover 3 days after implantation, and those that showed neurological deficits after surgery were sacrificed and excluded from statistical analyses.17,18 

Behavioral Studies

Tactile sensitivity was assessed by measuring the paw-withdrawal threshold of rats in response to probing with von Frey monofilaments (Stoelting, USA).19  Animals’ tactile thresholds before operation were set equal to 15 g.13  In some animals, the motor function was assessed using an accelerating rota-rod apparatus (LE8500; Ugo Basile, Italy). For acclimatization, animals were subjected to three training trials at 3- to 4-h intervals on 2 separate days. In training sessions, the rod was set to accelerate from 3 to 30 rpm over a 180-s period. In the test session, the performance times of rats were recorded up to a cutoff time of 180 s. Three measurements tested at intervals of 5 min were averaged in each test.20 

Western Blotting

The dissected dorsal horn (L4 to L5) sample was homogenized in 25 mM Tris-HCl, 150 mM NaCl, 1% NP-40, 1% sodium deoxycholate, and 0.1% sodium dodecyl sulfate with a complete protease inhibitor mixture (Roche, Germany). After incubation on ice (1 h), the lysates were centrifuged (14,000g, 20 min, 4°C). The cytoplasmic, nuclear extracts, and membrane protein were prepared by extraction reagent kits (NE-PER and Mem-PER; Thermo Scientific, USA) according to the manufacturer’s instructions. Protein concentrations were determined using a bicinchoninic acid assay. In brief, the supernatant was separated on acrylamide gel and transferred to a polyvinylidene difluoride membrane and then incubated (1 h, room temperature) in either rabbit anti-total HDAC4 (tHDAC4, 1:1,000; Genetex, USA), rabbit anti- phosphorylated HDAC4 (pHDAC4, 1:1,000; Genetex), rabbit anti-t14-3-3β (1:1,000; Millipore, USA), rabbit anti-pSGK (1:1,000; Abcam, United Kingdom), rabbit anti-tSGK1 (1:800; Santa Cruz Biotechnology, USA), or mouse antiglyceraldehyde 3-phosphate dehydrogenase (GAPDH, 1:20,000; Novus, USA). Blots were washed and incubated (1 h, room temperature) in peroxidase-conjugated goat anti-rabbit immunoglobulin G (1:8,000; Jackson ImmunoResearch, USA) or goat anti-mouse immunoglobulin G (1:8,000; Jackson ImmunoResearch). Protein bands were visualized by using an enhanced chemiluminescence detection kit (ECL Plus; Millipore), and, then, densitometric analysis of the Western blot membranes was performed with Science Lab 2003 (Fuji, Japan).

Coprecipitation

Rabbit polyclonal antibody against tHDAC4, t14-3-3β, and tSGK1 was incubated (overnight, 4°C) with the extraction of the dorsal horn samples. The 1:1 slurry protein agarose suspension (Millipore) was added into that immunocomplex of protein, and the mixture was incubated (2 to 3 h, 4°C). Agarose beads were washed once with 1% (v/v) Triton X-100 in an immunoprecipitation buffer (50 mM Tris-Cl, pH 7.4, 5 mM EDTA, and 0.02% [w/v] sodium azide), twice with 1% (v/v) Triton X-100 in an immunoprecipitation buffer plus 300 mM NaCl, and thrice with an immunoprecipitation buffer only. Binding proteins were eluted with sodium dodecyl sulfate polyacrylamide gel electrophoresis sample buffer at 95°C. Proteins were separated with the use of sodium dodecyl sulfate polyacrylamide gel electrophoresis, transferred to polyvinylidene difluoride membranes electrophoretically, and detected by using rabbit anti-tHDAC4 (1:1,000; Genetex), rabbit anti-pHDAC4 (1:1,000; GeneTex), rabbit anti-t14-3-3β (1:1,000; Millipore), rabbit anti-pSGK (1:1,000; Abcam), and rabbit anti-tSGK1 (1:800; Santa Cruz Biotechnology).

Immunofluorescence

After perfusion (100 ml phosphate-buffered saline followed by 300 ml paraformaldehyde, 4%; pH 7.4), the spinal cord samples were harvested, postfixed (4°C for 4 h), and cryoprotected in sucrose solution (30%) overnight. For double-labeling analyses investigating interactions between HDAC4 and neuronal/glial/microglia markers, the spinal sections were incubated with a mixture of rabbit anti-tHDAC4 (1:200; GeneTex) and mouse monoclonal antineuronal nuclear antigen (NeuN, a neuronal marker, 1:500; Millipore Bioscience Research Reagents), mouse antiglial fibrillary acidic protein (a marker of astroglial cells; 1:1,000; Millipore), or mouse anti-integrin αM (OX-42, a marker of microglia; 1:500; Santa Cruz Biotechnology) overnight (4°C). Then the sections were then incubated (1 h, 37°C) with Alexa Fluor 488 (1:1,500) and Alexa Fluor 594 (1:1,500; Invitrogen). When examining the interaction between HDAC4, 14-3-3β, and SGK1, the specific antibodies were mixed with 10X reaction buffer (Mix-n-Stain; Biotium, USA) with the antibody solution at a ratio of 1:10. Then, the solution was transferred to the viral containing dye (CF; Biotium) and incubated in the dark (30 min, at room temperature). The spinal cord sections were sequentially incubated (overnight, 4°C) with dilute solutions, that is, rabbit-pHDAC4 (200X; GeneTex), rabbit anti-t14-3-3β (×1000; Millipore), and rabbit anti-pSGK (×1,000; Abcam), and were washed five times in between each incubation.

Small-interfering RNA

The 19 nucleotide duplexes of the small-interfering RNAs (siRNAs) for 14-3-3β were 5’-GAAAUACAAUUCUGUUGUA-3’ and missense nucleotides were 5’-UACAACAGAAUUGUAUUUC-3’. The missense or siRNA with polyethyleneimine (10 μl, Al 25 kDa; Sigma–Aldrich, Germany)-based gene delivery system was intrathecally into the dorsal subarachnoid space (L4 to L5) of animals through the implanted catheter (daily for 4 days).

Drugs Application

GSK-650394 (a SGK1 inhibitor; 100 nM, 10 μl; Tocris Bioscience, USA) or LMK235 (an HDAC4 inhibitor; 10, 30, and 100 nM, 10 μl; Tocris Bioscience) was administered intrathecally by bolus injection. A vehicle solution of a volume identical to that of the tested agents was dispensed to serve as a control.

Data Analysis

All the data in this study were analyzed using SigmaPlot 10.0 (Systat Software, U.S.A.) or Prism 6.0 (GraphPad, U.S.A.) and are expressed as the mean ± SEM. Paired two-tailed Student t test was used to compare the means between groups. One-way or two-way ANOVAs were used to assess the changes in values for serial measurements over time, and post hoc Tukey tests were used to compare the means of groups. Significance was set at P value less than 0.05.

Spinal Nerve Ligation Induced Allodynia-associated Cytoplasmic HDAC4 Retention

Western blot analysis of dorsal horn samples (L4 to L5) demonstrated that the amount of the total HDAC4 protein (fig. 2A; tHDAC4) was uniformly expressed at days 1, 3, 7, 14, and 21 after operation in the ipsilateral (I) and contralateral (C) dorsal horn of both the spinal nerve ligation (SNL; all P > 0.05 compared with day 1, n = 6) and sham operation (Sham; all P > 0.05 compared with day 1, n = 6) groups. Nevertheless, when compared with the sham operation, SNL significantly increased the cytoplasm/nuclear ratio of tHDAC4 by increasing the abundance of the cytoplasmic but decreasing that of the nuclear HDAC4 in the ipsilateral (fig. 2B; P < 0.001, P < 0.001, P = 0.005, and P = 0.006, respectively compared with day 1, n = 6), but not the contralateral (all P > 0.05 compared with day 1, n = 6), dorsal horn at days 3, 7, 14, and 21 after operation, implying SNL-provoked cytoplasmic HDAC4 accumulation in the ipsilateral dorsal horn. The SNL-provoked HDAC4 retention was parallel to the time course of decrements in withdrawal latency, which occurred at days 3, 7, 14, and 21 after operation (fig. 2C; all P < 0.001 compared with day 1, n = 7). These results suggest that SNL-induced behavioral allodynia is accompanied with HDAC4 translocation in the dorsal horn.

Fig. 2.

Neuropathic injury induces behavioral allodynia and cytoplasmic histone deacetylase (HDAC4) retention in dorsal horn neurons. (A) Representative Western blots and statistical analysis showing total HDAC4 (tHDAC4) were uniformly expressed at days 1, 3, 7, 14, and 21 after operation in the ipsilateral (I, IPSI) and contralateral (C, CONTRA) dorsal horn of the spinal nerve ligation (SNL) and sham operation (Sham) groups (P > 0.05 compared with day 1, n = 6). (B) SNL statistically increased the tHDAC4 abundance in the cytoplasmic fraction but decreased that in the nuclear fractions of the ipsilateral dorsal horn samples at days 3, 7, 14, and 21 after operation that resulted in an increased cytoplasmic/nuclear ratio of tHDAC4 (*P < 0.05, **P < 0.01 compared with Sham IPSI; ##P < 0.01 compared with day 1, n = 6). (C) Von Frey test showing SNL significantly decreased the mechanical threshold of the ipsilateral hind paw at days 1, 3, 7, 14, and 21 after operation (**P < 0.01 compared with Sham IPSI; ##P < 0.01 compared with day 1, n = 7). (D) Immunohistochemistry images showing the tHDAC4 immunoreactivity (I; red) was colabeled (yellow) with NeuN (II; green, a marker for neurons), but not OX-42 (III; green, a marker for microglia) or glial fibrillary acidic protein (IV GFAP; green, a marker for astrocytes), in the ipsilateral dorsal horn of spinal cord sections obtained at day 7 after operation (SNL7D). Scale bar = 50 μm, thickness = 50 μm. Each of these images was replicated in seven sample preparations with similar results each time. GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting.

Fig. 2.

Neuropathic injury induces behavioral allodynia and cytoplasmic histone deacetylase (HDAC4) retention in dorsal horn neurons. (A) Representative Western blots and statistical analysis showing total HDAC4 (tHDAC4) were uniformly expressed at days 1, 3, 7, 14, and 21 after operation in the ipsilateral (I, IPSI) and contralateral (C, CONTRA) dorsal horn of the spinal nerve ligation (SNL) and sham operation (Sham) groups (P > 0.05 compared with day 1, n = 6). (B) SNL statistically increased the tHDAC4 abundance in the cytoplasmic fraction but decreased that in the nuclear fractions of the ipsilateral dorsal horn samples at days 3, 7, 14, and 21 after operation that resulted in an increased cytoplasmic/nuclear ratio of tHDAC4 (*P < 0.05, **P < 0.01 compared with Sham IPSI; ##P < 0.01 compared with day 1, n = 6). (C) Von Frey test showing SNL significantly decreased the mechanical threshold of the ipsilateral hind paw at days 1, 3, 7, 14, and 21 after operation (**P < 0.01 compared with Sham IPSI; ##P < 0.01 compared with day 1, n = 7). (D) Immunohistochemistry images showing the tHDAC4 immunoreactivity (I; red) was colabeled (yellow) with NeuN (II; green, a marker for neurons), but not OX-42 (III; green, a marker for microglia) or glial fibrillary acidic protein (IV GFAP; green, a marker for astrocytes), in the ipsilateral dorsal horn of spinal cord sections obtained at day 7 after operation (SNL7D). Scale bar = 50 μm, thickness = 50 μm. Each of these images was replicated in seven sample preparations with similar results each time. GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting.

Close modal

Spinal HDAC4 Was Localized in Dorsal Horn Neurons

Double-staining immunohistochemistry images revealed that most of the HDAC4 immunofluorescence (fig. 2D; I tHDAC4) occurred coincidently with NeuN (II, a neuronal marker), instead of OX-42 (III, a microglial marker) or glial fibrillary acidic protein (IV, an astrocyte marker) immunofluorescence, in the ipsilateral dorsal horn (L4 to L5) dissected at day 7 after SNL, suggesting that the neuropathic injury–associated spinal HDAC4 was present in the dorsal horn neurons but rather than in microglia or astrocytes.

SNL Induced HDAC4 Phosphorylation and Coupling with 14-3-3β

Spinal nerve ligation significantly increased the abundance of the phosphorylated HDAC4 (fig. 3A; pHDAC4) in the ipsilateral (I; P = 0.003, P = 0.004, P = 0.006, and P = 0.009, respectively, compared with day 1, n = 6), but not the contralateral (C; all P > 0.05 compared with day 1, n = 6), dorsal horn sample at days 3, 7, 14, and 21 after operation. SNL also up-regulated the expression of total 14-3-3β (fig. 3B; t14-3-3β) selectively in the ipsilateral dorsal horn at these time points (P = 0.005, P < 0.001, P = 0.001, and P < 0.001, respectively, compared with day 1, n = 6). However, the sham operation exhibited no effect in the abundance of pHDAC or t14-3-3β at days 3, 7, 14, and 21 after operation (fig. 3, A and B; all P > 0.05 compared with day 1, n = 6). When compared with the preoperation control (fig. 3C; day 1), SNL increased the amounts of tHDAC4-bound pHDAC4 and t14-3-3β in the tHDAC4-recognized precipitates at days 3 and 7 after operation (approximately 1.8- and 2.4-fold above day 1 in pHDAC4 and 1.4- and 1.6-fold above day 1 in t14-3-3β, respectively). The amount of t14-3-3β-bound pHDAC4 also was enhanced at the same time points in the 14-3-3β-recognized precipitates (approximately 2.0- and 2.8-fold above day 1). Nevertheless, the abundance of GAPDH in the preprecipitated homogenates obtained at these time points remained at a relative constant level. At day 7 after operation, SNL notably enhanced the immunofluorescence of pHDAC4 (fig. 4A; I and IV pHDAC4) and total 14-3-3β (II and V t14-3-3β) in the ipsilateral (IPSI) compared with the contralateral dorsal horn (CONTRA I and VII pHDAC4; II and VIII t14-3-3β). Merged images showed that most of the pHDAC4 immunoreactivity was coincident with that of 14-3-3β (III, VI, and VIa) in the ipsilateral dorsal horn. Statistical analysis revealed that SNL significantly enhanced the numbers of pHDAC4-, t14-3-3β-, and pHDAC4-t14-3-3β double-labeled neurons in the ipsilateral compared with the contralateral dorsal horn (fig. 4B; all P < 0.01 compared with CONTRA, n = 7). Together, these results showed that SNL provoked HDAC4 phosphorylation, 14-3-3β expression, HDAC4-14-3-3β coupling/colocalization, and HDAC4 translocation in dorsal horn neurons that have a temporal correlation with behavioral allodynia.

Fig. 3.

Neuropathic injury provokes spinal phosphorylated histone deacetylase (pHDAC4) and 14-3-3β expressions and HDAC4-14-3-3β coupling. (A and B) Representative Western blots and statistical analysis showing, when compared with the sham operation (Sham), spinal nerve ligation (SNL) statistically up-regulated the level of pHDAC4 and total 14-3-3β (t14-3-3β) at days 3, 7, 14, and 21 after operation in the ipsilateral (I, IPSI) but not the contralateral (C, CONTRA) dorsal horn (**P < 0.01 compared with the Sham IPSI; ##P < 0.01 compared with day 1, n = 6). (C) Coimmunoprecipitation analysis showing SNL enhanced the tHDAC4-bound pHDAC4 and t14-3-3β- and t14-3-3β-bound pHDAC4 in the dorsal horn samples at days 3 and 7 after operation. In both cases, no detectable immunoreactivity was labeled by antibodies in the control IgG-recognized precipitation. GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting; IgG = immunoglobulin G; In = input control; IP = immunoprecipitation.

Fig. 3.

Neuropathic injury provokes spinal phosphorylated histone deacetylase (pHDAC4) and 14-3-3β expressions and HDAC4-14-3-3β coupling. (A and B) Representative Western blots and statistical analysis showing, when compared with the sham operation (Sham), spinal nerve ligation (SNL) statistically up-regulated the level of pHDAC4 and total 14-3-3β (t14-3-3β) at days 3, 7, 14, and 21 after operation in the ipsilateral (I, IPSI) but not the contralateral (C, CONTRA) dorsal horn (**P < 0.01 compared with the Sham IPSI; ##P < 0.01 compared with day 1, n = 6). (C) Coimmunoprecipitation analysis showing SNL enhanced the tHDAC4-bound pHDAC4 and t14-3-3β- and t14-3-3β-bound pHDAC4 in the dorsal horn samples at days 3 and 7 after operation. In both cases, no detectable immunoreactivity was labeled by antibodies in the control IgG-recognized precipitation. GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting; IgG = immunoglobulin G; In = input control; IP = immunoprecipitation.

Close modal
Fig. 4.

Neuropathic injury up-regulates colocalized phosphorylated histone deacetylase (pHDAC4) and t14-3-3β expressions in the ipsilateral dorsal horn. (A) Immunohistochemical images showing that spinal nerve ligation (SNL) enhanced the immunoreactivity of pHDAC4 (I and IV, red) and total 14-3-3β (t14-3-3β; II and V, green) in the ipsilateral (IPSI) compared with the contralateral (CONTRA) dorsal horn (I, VII and II, VIII, respectively) of the spinal sections obtained at day 7 after operation (7D). Overlay images and the count of double-labeled neurons showing the enhanced pHDAC4 fluorescence (IV) costained with t14-3-3β immunoreactivity (V) in the ipsilateral (III, VI, and VIa, yellow), but not in the contralateral (III, IX, and IXa), dorsal horn. Scale bar = 50 μm, thickness = 50 μm. Each of these immunofluorescence images was replicated in seven sample preparations with similar results each time. (B) Statistical analysis showing SNL enhanced the count of pHDAC4-positive, t14-3-3β-positive, and double-labeled neurons in the ipsilateral dorsal horn of the spinal sections obtained at day 7 after operation (**P < 0.01 compared with the CONTRA, n = 7).

Fig. 4.

Neuropathic injury up-regulates colocalized phosphorylated histone deacetylase (pHDAC4) and t14-3-3β expressions in the ipsilateral dorsal horn. (A) Immunohistochemical images showing that spinal nerve ligation (SNL) enhanced the immunoreactivity of pHDAC4 (I and IV, red) and total 14-3-3β (t14-3-3β; II and V, green) in the ipsilateral (IPSI) compared with the contralateral (CONTRA) dorsal horn (I, VII and II, VIII, respectively) of the spinal sections obtained at day 7 after operation (7D). Overlay images and the count of double-labeled neurons showing the enhanced pHDAC4 fluorescence (IV) costained with t14-3-3β immunoreactivity (V) in the ipsilateral (III, VI, and VIa, yellow), but not in the contralateral (III, IX, and IXa), dorsal horn. Scale bar = 50 μm, thickness = 50 μm. Each of these immunofluorescence images was replicated in seven sample preparations with similar results each time. (B) Statistical analysis showing SNL enhanced the count of pHDAC4-positive, t14-3-3β-positive, and double-labeled neurons in the ipsilateral dorsal horn of the spinal sections obtained at day 7 after operation (**P < 0.01 compared with the CONTRA, n = 7).

Close modal

14-3-3β Knockdown Ameliorated Allodynia, HDAC4 Translocation, and HDAC4-14-3-3β Coupling

Although exhibiting no effect on the level of GAPDH, the administration with 14-3-3β messenger RNA (mRNA)- targeting siRNA (fig. 5A; 14-3-3β messenger RNA-targeting small interfering RNA (14-3-3β RNAi); 10 μl, P = 0.243, P < 0.001, and P < 0.001 in 1, 3, and 5 μg, respectively, compared with naive, n = 6), but rather than the administration with missense siRNA (14-3-3β MS; 5 μg, 10 μl), administration with polyethylenimine (a transfection reagent; 10 μl), or implantation of intrathecal catheter (it), dose dependently decreased the abundance of t14-3-3β in the dorsal horn samples when compared with the control levels in naive animals (Naive; all P > 0.05 compared with naive, n = 6), indicating specific siRNA sufficiently knocked-down spinal 14-3-3β expression. No statistical difference was evidenced in the performance time of the rota-rod test between the naive and polyethylenimine-, missense siRNA-, or 14-3-3β mRNA-targeting siRNA (3 μg, 10 μl)-treated animals (fig. 5B; all P > 0.05 compared with day 0, n = 7), indicating that our knockdown procedures resulted in no motor deficits in animals. All the treatments, including catheter implantation and antisense or missense siRNA injection, exhibited no difference in the withdrawal threshold of von Frey test measured in sham-operated animals (fig. 5C; Sham; all P > 0.05 compared with Sham, n = 7). In contrast, the spinal administration with 14-3-3β mRNA-targeting siRNA (fig. 5D; SNL + it + 14-3-3β RNAi; P = 0.001 and P < 0.001, respectively, compared with SNL, n = 7), but not the administration with missense siRNA (SNL + it + 14-3-3β MS) or catheter implantation alone (SNL + it), partially ameliorated behavioral allodynia as evidenced by a significant increase in the withdrawal threshold at days 5 and 7 after operation compared with animals that received SNL only (SNL; all P > 0.05 compared with SNL, n = 7), indicating that loss of dorsal horn 14-3-3β alleviated SNL-associated allodynia. Although both treatments failed to affect the SNL-enhanced pHDAC4 level (fig. 5E; all P > 0.05 compared with SNL 7D, n = 6), daily administration with 14-3-3β mRNA-targeting siRNA (fig. 5F; SNL 7D + 14-3-3β RNAi; P = 0.015 compared with SNL 7D, n = 6), but rather than the missense siRNA (SNL 7D + 14-3-3β MS; P > 0.05 compared with SNL 7D, n = 6), attenuated the SNL-provoked cytoplasmic HDAC4 retention in the ipsilateral dorsal horn when compared with the SNL group. Administering with 14-3-3β mRNA-targeting siRNA reduced the amount of tHDAC4-bound t14-3-3β in the ipsilateral dorsal horn measured at day 7 after nerve ligation (approximately 0.6-fold above SNL 7D) when compared with animals that received no siRNA (SNL 7D; approximately 1.9-fold above Sham 7D). Nevertheless, it failed to affect SNL-enhanced abundance of tHDAC4-bound pHDAC4 (approximately 1.1-fold above SNL 7D; fig. 5G). Together with the above results, these data revealed that without affecting HDAC4 phosphorylation, the 14-3-3β knockdown ameliorated SNL-induced allodynia as well as the HDAC4-14-3-3β coupling and cytoplasmic HDAC4 accumulation.

Fig. 5.

14-3-3β small-interfering RNA (siRNA) prevents neuropathic injury–associated allodynia, histone deacetylase (HDAC4) translocation, and HDAC4-14-3-3β coupling. (A) Representative Western blot and statistical analysis showing administration with 14-3-3β messenger RNA-targeting small interfering RNA (14-3-3β RNAi), but not intrathecal catheter implantation (it), administration with polyethylenimine (PEI; a transfection reagent; 10 μl), or missense siRNA (14-3-3β MS, 5 μg, 10 μl), dose dependently decreased the level of total 14-3-3β (t14-3-3β) in the dorsal horn (**P < 0.01 compared with the Naive, n = 6). (B) Administration with PEI, missense siRNA, and 14-3-3β mRNA-targeting siRNA (3 μg, 10 μl) failed to affect the performance times of rata-rod test at days 0, 1, 2, 3, and 4 after treatment compared with the naive animals (P > 0.05 compared with naive, n = 7). (C and D) Although treatments failed to produce statistical differences in sham-operated animals (Sham; P > 0.05 compared with Sham, n = 7), 14-3-3β RNA-targeting siRNA significantly increased the withdrawal threshold in spinal nerve ligation (SNL) animals at days 5 and 7 after operation (**P < 0.01 compared with the SNL, n = 7). (BD) The gray bar at the bottom indicates the duration of administration. (E and F) Representative Western blot and statistical analysis showing while 14-3-3β mRNA-targeting siRNA failed to affect the pHDAC4 expression, it significantly decreased the SNL-enhanced cytoplasmic/nuclear tHDAC4 ratio (*P < 0.05, **P < 0.01 compared with Sham 7D, #P < 0.05 compared with SNL 7D, n = 6). (G) 14-3-3β mRNA-targeting siRNA reversed the SNL-increased tHDAC4-t14-3-3β but rather than tHDAC4-pHDAC4 coprecipitation. No detectable immunoreactivity was labeled by antibodies in the control IgG-recognized precipitation. GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting; IgG = immunoglobulin G; In = input control; IP = immunoprecipitation.

Fig. 5.

14-3-3β small-interfering RNA (siRNA) prevents neuropathic injury–associated allodynia, histone deacetylase (HDAC4) translocation, and HDAC4-14-3-3β coupling. (A) Representative Western blot and statistical analysis showing administration with 14-3-3β messenger RNA-targeting small interfering RNA (14-3-3β RNAi), but not intrathecal catheter implantation (it), administration with polyethylenimine (PEI; a transfection reagent; 10 μl), or missense siRNA (14-3-3β MS, 5 μg, 10 μl), dose dependently decreased the level of total 14-3-3β (t14-3-3β) in the dorsal horn (**P < 0.01 compared with the Naive, n = 6). (B) Administration with PEI, missense siRNA, and 14-3-3β mRNA-targeting siRNA (3 μg, 10 μl) failed to affect the performance times of rata-rod test at days 0, 1, 2, 3, and 4 after treatment compared with the naive animals (P > 0.05 compared with naive, n = 7). (C and D) Although treatments failed to produce statistical differences in sham-operated animals (Sham; P > 0.05 compared with Sham, n = 7), 14-3-3β RNA-targeting siRNA significantly increased the withdrawal threshold in spinal nerve ligation (SNL) animals at days 5 and 7 after operation (**P < 0.01 compared with the SNL, n = 7). (BD) The gray bar at the bottom indicates the duration of administration. (E and F) Representative Western blot and statistical analysis showing while 14-3-3β mRNA-targeting siRNA failed to affect the pHDAC4 expression, it significantly decreased the SNL-enhanced cytoplasmic/nuclear tHDAC4 ratio (*P < 0.05, **P < 0.01 compared with Sham 7D, #P < 0.05 compared with SNL 7D, n = 6). (G) 14-3-3β mRNA-targeting siRNA reversed the SNL-increased tHDAC4-t14-3-3β but rather than tHDAC4-pHDAC4 coprecipitation. No detectable immunoreactivity was labeled by antibodies in the control IgG-recognized precipitation. GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting; IgG = immunoglobulin G; In = input control; IP = immunoprecipitation.

Close modal

GSK-650394 Prevented Allodynia, HDAC4-14-3-3β Coupling, and HDAC4 Phosphorylation/Translocation

A bolus injection of GSK-650394 (fig. 6A; SNL 7D + GSK; an SGK1 inhibitor; 100 nM, 10 μl: P < 0.001, P < 0.001, P = 0.001, P = 0.01, and P = 0.01, respectively, compared with hour 0, n = 7), but not the vehicle solution (SNL 7D + Veh; all P > 0.05 compared with hour 0, n = 7), significantly alleviated SNL-induced allodynia at hours 1, 2, 3, 4, and 5 after administration on day 7 after SNL when compared with animals subjected to SNL only. This result indicates that antagonism of spinal SGK1 alleviated neuropathic injury–established nociception enhancement. Although the intrathecal GSK-650394 injection significantly reversed the increases in the pHDAC4 amount (fig. 6B; SNL 7D + GSK; P = 0.015 compared with SNL 7D, n = 6) and the cytoplasmic/nuclear ratio of tHDAC4 (fig. 6C; SNL 7D + GSK; P < 0.001 compared with SNL 7D, n = 6), it failed to affect the SNL-enhanced 14-3-3β expression (fig. 6D; SNL 7D + GSK; P > 0.05 compared with SNL 7D, n = 6) in the ipsilateral dorsal horn. The SNL-enhanced amounts of tSGK1-bound pSGK1, pHDAC4, and t14-3-3β (approximately 1.8-, 2.4- and 2.0-fold above Sham 7D) as well as tHDAC4-bound pSGK1, pHDAC4, and t14-3-3β (approximately 2.0-, 1.8-, and 2.2-fold above Sham 7D) in the ipsilateral dorsal horn were reduced by the spinal GSK-650394 injection (approximately 0.6-, 0.2- and 0.4-fold above SNL 7D in tSGK1 and 0.7-, 0.5-, and 0.4-fold above SNL 7D in tHDAC4, respectively). Although daily administration with 14-3-3β mRNA-targeting siRNA (SNL 7D + 14-3-3β RNAi) decreased the tSGK1- and f4-bound 14-3-3β (approximately 0.2- and 0.3-fold above SNL 7D, respectively), it failed to affected the abundance of tSGK1-bound pSGK1 and pHDAC4 (approximately 1.4- and 1.4-fold above Sham 7D in pSGK1 and 2.2- and 2.3-fold above SNL 7D in pHDAC4, respectively) or tHDAC4-bound pSGK1 and pHDAC4 (approximately 1.1- and 1.0-fold above SNL 7D in pSGK1 and 1.0- and 0.9-fold above SNL 7D in pHDAC4, respectively) measured on day 7 postoperation (fig. 6E). Moreover, when compared with sham operation, SNL provoked dorsal horn SGK1 phosphorylation as evidenced by significantly increased amounts of phosphorylated SGK1 (fig. 6F; pSGK1; P < 0.001 compared with Sham 7D, n = 6) in the ipsilateral dorsal horn on day 7 after operation. In contrast to intrathecal GSK-650394 injection that effectively reversed the SNL-enhanced amount of pSGK1 (SNL 7D + GSK; P = 0.002 compared with SNL 7D, n = 6), treatments including the spinal injection of the vehicle solution (SNL 7D + Veh), the daily administration with 14-3-3β mRNA-targeting siRNA and missense siRNA (SNL 7D + 14-3-3β MS) failed to affect the SNL-associated increase in pSGK1 level (all P > 0.05 compared with SNL 7D, n = 6), indicating that the SNL-induced SGK1 phosphorylation and SGK1-HDAC4 interactions were not affected by focal knockdown of spinal 14-3-3β expression.

Fig. 6.

Neuropathic injury provokes serum- and glucocorticoid-inducible kinase 1 (SGK1)-dependent histone deacetylase (HDAC4) phosphorylation and HDAC4-14-3-3β association in dorsal horn neurons. (A) At day 7 (7D) after spinal nerve ligation (SNL), administration with GSK-650394 (an SGK1 inhibitor; 30 nM, 10 μl; SNL 7D + GSK) increased the withdrawal threshold of the ipsilateral hind paw at hours 1, 2, 3, 4, and 5 after treatment (**P < 0.01 compared with the hour 0, n = 7). (BD) Representative Western blot and statistical analysis showing, when compared with the sham operation (Sham 7D), that SNL enhanced the amount of phosphorylated HDAC4 (B, pHDAC4), the cytoplasmic/nuclear ratio of total HDAC4 (C, tHDAC4), and the amount of total 14-3-3β (D, t14-3-3β) in the dorsal horn (SNL 7D). Bolus of GSK-650394 decreased the SNL-enhanced pHDAC4 and cytoplasmic/nuclear ratio of tHDAC4 but exhibited no effect on the SNL-enhanced t14-3-3β abundance (*P < 0.05, **P < 0.01 compared with Sham 7D; #P < 0.05, ##P < 0.01 compared with the SNL 7D, n = 6). (E) Coprecipitation analysis showing SNL notably increased the amounts of tSGK1-bound pSGK1, pHDAC4, and t14-3-3β and tHDAC4-bound pSGK1, pHDAC4 and t14-3-3β that were all significantly reduced by GSK-650394. Although administration with 14-3-3β messenger RNA-targeting small-interfering RNA (siRNA; SNL 7D + 14-3-3β RNAi) decreased the SNL-enhanced amount of tSGK1- and tHDAC4-bound 14-3-3β, it failed to affected that of tSGK1-bound pSGK1 and pHDAC4 or tHDAC4-bound pSGK1 and pHDAC4. No detectable immunoreactivity was labeled by antibodies in the control IgG-recognized precipitates in both cases. (F) Representative Western blot and statistical analysis showing administration with GSK-650394, but not the 14-3-3β mRNA-targeting siRNA, decreased the SNL-enhanced abundance of pSGK1 (**P < 0.01 compared with the Sham 7D, ##P < 0.01 compared with the SNL 7D, n = 6). GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting; IgG = immunoglobulin G; In = input control; IP = immunoprecipitation; Veh = vehicle solution.

Fig. 6.

Neuropathic injury provokes serum- and glucocorticoid-inducible kinase 1 (SGK1)-dependent histone deacetylase (HDAC4) phosphorylation and HDAC4-14-3-3β association in dorsal horn neurons. (A) At day 7 (7D) after spinal nerve ligation (SNL), administration with GSK-650394 (an SGK1 inhibitor; 30 nM, 10 μl; SNL 7D + GSK) increased the withdrawal threshold of the ipsilateral hind paw at hours 1, 2, 3, 4, and 5 after treatment (**P < 0.01 compared with the hour 0, n = 7). (BD) Representative Western blot and statistical analysis showing, when compared with the sham operation (Sham 7D), that SNL enhanced the amount of phosphorylated HDAC4 (B, pHDAC4), the cytoplasmic/nuclear ratio of total HDAC4 (C, tHDAC4), and the amount of total 14-3-3β (D, t14-3-3β) in the dorsal horn (SNL 7D). Bolus of GSK-650394 decreased the SNL-enhanced pHDAC4 and cytoplasmic/nuclear ratio of tHDAC4 but exhibited no effect on the SNL-enhanced t14-3-3β abundance (*P < 0.05, **P < 0.01 compared with Sham 7D; #P < 0.05, ##P < 0.01 compared with the SNL 7D, n = 6). (E) Coprecipitation analysis showing SNL notably increased the amounts of tSGK1-bound pSGK1, pHDAC4, and t14-3-3β and tHDAC4-bound pSGK1, pHDAC4 and t14-3-3β that were all significantly reduced by GSK-650394. Although administration with 14-3-3β messenger RNA-targeting small-interfering RNA (siRNA; SNL 7D + 14-3-3β RNAi) decreased the SNL-enhanced amount of tSGK1- and tHDAC4-bound 14-3-3β, it failed to affected that of tSGK1-bound pSGK1 and pHDAC4 or tHDAC4-bound pSGK1 and pHDAC4. No detectable immunoreactivity was labeled by antibodies in the control IgG-recognized precipitates in both cases. (F) Representative Western blot and statistical analysis showing administration with GSK-650394, but not the 14-3-3β mRNA-targeting siRNA, decreased the SNL-enhanced abundance of pSGK1 (**P < 0.01 compared with the Sham 7D, ##P < 0.01 compared with the SNL 7D, n = 6). GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting; IgG = immunoglobulin G; In = input control; IP = immunoprecipitation; Veh = vehicle solution.

Close modal

14-3-3β Knockdown Failed to Affect SGK1-HDAC4 Colocalization

When compared with sham-operated animals (fig. 7A; Sham I and II), SNL notably enhanced the immunoreactivity of pSGK1 and pHDAC4 in the ipsilateral dorsal horn (IV and V). In addition, pSGK1 and pHDAC4 were largely shown to colocalize in the merged image (VI and VIa). Intrathecal GSK-650394 injection markedly reversed the SNL-enhanced pSGK1, pHDAC4, and colocalized immunoreactivity in the dorsal horn (VII, VIII, IX, and IXa), whereas daily 14-3-3β mRNA-targeting siRNA (X, XI, XII, and XIIa) had no effect. Statistical analysis revealed, when compared with sham operation, that SNL significantly enhanced the counts of pSGK1-, pHDAC4-, and pSGK1-pHDAC4 double-labeled neurons in the ipsilateral dorsal horn (fig. 7B; all P < 0.001 compared with Sham 7D, n = 6) that were reversed by administering with a bolus of GSK-650394 (P = 0.002, P < 0.001, and P < 0.001, respectively, compared with SNL 7D, n = 6) but rather than daily 14-3-3β mRNA-targeting siRNA (all P > 0.05 compared with SNL 7D, n = 6). These data indicated that the SNL-provoked SGK1 and HDAC4 phosphorylation and SGK1-HDAC4 colocalization were not affected by focal knockdown of spinal 14-3-3β expression.

Fig. 7.

GSK-650394, but not 14-3-3β small-interfering RNA (siRNA), prevented neuropathic injury–induced phosphorylated serum- and glucocorticoid-inducible kinase 1 (pSGK1)-phosphorylated histone deacetylase (pHDAC4) colocalization. (A) Immunohistochemical images showing spinal nerve ligation (SNL) notably enhanced the immunoreactivity of pSGK1 (IV, green) and pHDAC4 (V, red) compared with the sham group (Sham, I and II, respectively) in the ipsilateral dorsal horn of spinal slices obtained at day 7 after operation (7D). Overlay images and the count of double-labeled neurons show SNL enhanced the colocalized immunoreactivity of pSGK1 with pHDAC4 in the dorsal horn (VI and VIa, yellow), which were remarkably reduced by the administration with GSK-650394 (an SGK1 inhibitor; 30 nM, 10 μl; SNL7D + GSK, IX and IXa), but not 14-3-3β messenger RNA-targeting small interfering RNA (SNL 7D + 14-3-3β RNAi, XII and XIIa). (B) Statistical analysis showing SNL notably enhanced the count of pSGK1-positive, pHDAC4-positive, and double-labeled dorsal horn that were remarkably reduced by the administration with GSK-650394 (SNL7D + GSK), but not 14-3-3β mRNA-targeting siRNA (SNL 7D + 14-3-3β RNAi, ** P < 0.01 compared with the Sham 7D, ##P < 0.01 compared with the SNL 7D, n = 7).

Fig. 7.

GSK-650394, but not 14-3-3β small-interfering RNA (siRNA), prevented neuropathic injury–induced phosphorylated serum- and glucocorticoid-inducible kinase 1 (pSGK1)-phosphorylated histone deacetylase (pHDAC4) colocalization. (A) Immunohistochemical images showing spinal nerve ligation (SNL) notably enhanced the immunoreactivity of pSGK1 (IV, green) and pHDAC4 (V, red) compared with the sham group (Sham, I and II, respectively) in the ipsilateral dorsal horn of spinal slices obtained at day 7 after operation (7D). Overlay images and the count of double-labeled neurons show SNL enhanced the colocalized immunoreactivity of pSGK1 with pHDAC4 in the dorsal horn (VI and VIa, yellow), which were remarkably reduced by the administration with GSK-650394 (an SGK1 inhibitor; 30 nM, 10 μl; SNL7D + GSK, IX and IXa), but not 14-3-3β messenger RNA-targeting small interfering RNA (SNL 7D + 14-3-3β RNAi, XII and XIIa). (B) Statistical analysis showing SNL notably enhanced the count of pSGK1-positive, pHDAC4-positive, and double-labeled dorsal horn that were remarkably reduced by the administration with GSK-650394 (SNL7D + GSK), but not 14-3-3β mRNA-targeting siRNA (SNL 7D + 14-3-3β RNAi, ** P < 0.01 compared with the Sham 7D, ##P < 0.01 compared with the SNL 7D, n = 7).

Close modal

LMK235 Prevented Allodynia but Not SGK1 Phosphorylation, HDAC4 Phosphorylation/Translocation, and 14-3-3β Expression

On day 7 after SNL, a bolus injection of LMK235 (fig. 8A; SNL 7D + LMK235; an HDAC4 inhibitor; 10 μl; P = 0.456, P = 0.002, P = 0.005, and P = 0.112; P = 0.005, P < 0.001, P = 0.001, and P < 0.001; all P < 0.001 in 10, 30, and 100 nM, respectively, compared with hour 0, n = 7), but not the vehicle solution (SNL 7D + Veh; all P > 0.05 compared with hour 0, n = 7), dose dependently alleviated SNL-induced allodynia at hours 2, 3, 4, and 5 after administration when compared with animals subjected to SNL only, indicating that inhibition of HDAC4 activity can alleviate neuropathic injury–established nociception enhancement. Nevertheless, we observe that intrathecal LMK235 injection (fig. 8, B and C; SNL 7D + LMK235; 100 nM, 10 μl) exhibited no effect on the SNL-enhanced t14-3-3β, pSGK1, pHDAC4 expressions and cytoplasmic/nuclear ratio of tHDAC4 (all P > 0.05 compared with SNL 7D, n = 6), indicating that the inhibition of HDAC4 activity by LMK235 might ameliorate SNL-induced allodynia through pathways other than the spinal SGK1/HDAC4/14-3-3β cascade.

Fig. 8.

LMK235 prevented allodynia but not serum- and glucocorticoid-inducible kinase 1 (SGK1) phosphorylation, histone deacetylase (HDAC4) phosphorylation/translocation, and 14-3-3β expression. (A) At day 7 (7D) after spinal nerve ligation (SNL), administration with LMK235 (an HDAC4 and 5 inhibitor) increased the withdrawal threshold of the ipsilateral hind paw at hours 2, 3, 4, and 5 after treatment (**P < 0.01 compared with hour 0, n = 7). (B and C) Representative Western blot and statistical analysis showing LKM235 exhibited no effect on the SNL-enhanced amount of total 14-3-3β (t14-3-3β), phosphorylated SGK1 (pSGK1), phosphorylated HDAC4 (pHDAC4), and the cytoplasmic/nuclear ratio of total HDAC4 (*P < 0.05, **P < 0.01 compared with the sham operation [Sham] 7D, n = 6). GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting; Veh = vehicle solution.

Fig. 8.

LMK235 prevented allodynia but not serum- and glucocorticoid-inducible kinase 1 (SGK1) phosphorylation, histone deacetylase (HDAC4) phosphorylation/translocation, and 14-3-3β expression. (A) At day 7 (7D) after spinal nerve ligation (SNL), administration with LMK235 (an HDAC4 and 5 inhibitor) increased the withdrawal threshold of the ipsilateral hind paw at hours 2, 3, 4, and 5 after treatment (**P < 0.01 compared with hour 0, n = 7). (B and C) Representative Western blot and statistical analysis showing LKM235 exhibited no effect on the SNL-enhanced amount of total 14-3-3β (t14-3-3β), phosphorylated SGK1 (pSGK1), phosphorylated HDAC4 (pHDAC4), and the cytoplasmic/nuclear ratio of total HDAC4 (*P < 0.05, **P < 0.01 compared with the sham operation [Sham] 7D, n = 6). GADPH = glyceraldehyde 3-phosphate dehydrogenase; IB = immunoblotting; Veh = vehicle solution.

Close modal

Results in this study demonstrated that neuropathic injury–associated nociception hypersensitivity involves spinal SGK1 activation–dependent HDAC4 phosphorylation and the subsequent HDAC4-14-3-3β coupling, which restricts HDAC4 location to the cytoplasm of dorsal horn neurons (fig. 1). We speculate that cytoplasmic HDAC4 retention, which reduces the amount of nucleus-bound HDAC4, could alter nociception-associated gene expression via increasing the histone acetylation–dependent chromatin remodeling. Although intrathecally administrated animals with an HDAC4 inhibitor effectively prevented SNL-induced allodynia, it failed to affect the associated HDAC4 phosphorylation. Although mechanisms underlying such a discrepancy are unclear, we suggest that the SGK1-dependent HDAC4 phosphorylation/translocation could contribute to the SNL-enhanced nociception through pathways other than the traditional HDAC4-associated epigenetic modulation, which increases histone acetylation to regulate chromatin remodeling. Our speculation was based on crystallography studies documented HDAC4 has three serine residues at amino acid 246, 467, and 632 that can be phosphorylated and a regulatory zinc-binding domain at amino acid 648 to 675 that allows binding to the transcriptional corepressor.21  Several HDAC inhibitors have been shown to act as a chelator for the zinc ion in the active site to hamper the association of HDAC with corepressors that impedes HDAC-mediated epigenic modification,22  and administration of HDAC4 inhibitors could have little effect on HDAC4 phosphorylation. Our proposal was supported by a study showing that class IIa HDAC inhibition, MC1568 or TMP269, does not prevent agonist-induced HDAC4 phosphorylation and nuclear extrusion of HDAC4 in cultured cells.23  Nevertheless, further experiments are needed to clarify the detailed mechanism.

In the current study, GSK-650394 administration alleviated SNL-induced allodynia as early as 1 h postinjection. This latency does not seem long enough for the recognized epigenetic expression of nociception-associated genes. It worth notice that SGK1 was demonstrated to phosphorylate Nedd4-2 and thus facilitate 14-3-3-Nedd4-2 coupling to regulate membrane trafficking of channel proteins in cultured cells.9  HDAC4 phosphorylation and subsequent coupling with 14-3-3β were also shown to regulate nucleocytoplasmic HDAC4 shuttling.7,11  Our previous study demonstrated SGK1 contributes to postinflammatory hyperalgesia by regulating the membrane trafficking of specific glutamatergic receptors, a pivotal spinal machinery underlying nociception hypersensitivity.13,18  Further experiments should be conducted to elucidate whether subcellular trafficking of glutamatergic receptors lies downstream of SGK1/pHDAC4/HDAC4-14-3-3β cascade to mediate neuropathic pain.

In this study, we showed that neuropathic injury provoked spinal SGK1-dependent HDAC4 phosphorylation and cytoplasmic retention. Nevertheless, the precise site where the SGK1-dependent HDAC4 phosphorylation occurs cannot be easily clarified, for which we analyzed the amount of pHDAC4 in dorsal horn homogenate. On the basis of the evidences demonstrated, HDAC4 phosphorylation promotes nuclear export but prevents nuclear import of HDAC4.24  Moreover, 14-3-3β functions as a cytoplasmic anchoring protein for HDAC4.25,26  We speculated that HDAC4 phosphorylation possibly occurs in the nucleus and the following association with 14-3-3β restricts HDAC4 location by anchoring this complex in the cytoplasm. Our speculation was derived from findings that the knockdown of 14-3-3β expression exhibited no effect on HDAC4 phosphorylation. However, it did attenuate cytoplasmic HDAC4 accumulation because there is no 14-3-3β available to anchor the HDAC4 shuttled from the nucleus after phosphorylation. Contrarily, if HDAC4 phosphorylation occurs within the cytoplasm, knockdown of 14-3-3β would not affect the amount of cytoplasmic HDAC4 because the degree of HDAC4 phosphorylation kept relatively constant. Our theory was supported by a study, in which either one or combinations of three HDAC4 shuttling–related residues were mutated, demonstrated HDAC4 is predominantly cytoplasmic in wild-type and single mutant cells, whereas it is nuclear in most triple mutant cells.27  Moreover, results obtained from cultured corticotrophinoma cells provides an indirect evidence by showing that most SGK1 was cytosolic before stimulation, whereas the amount of phosphorylated and nucleus-bound SGK1 increased after dexamethasone activation.28  We, therefore, propose that neuropathic injury provokes phosphorylation and subsequently nuclear accumulation of SGK1, which is followed by HDAC4 phosphorylation in the nucleus of dorsal horn neurons. However, the precise site where HDAC4 phosphorylation occurs requires further study to be clarified.

Similar to the study of Laffray et al.,10  in this study, we demonstrated that neuropathic injury enhanced the abundance of dorsal horn 14-3-3, a ubiquitous phosphor-binding protein.29  Conversely, trim-down of spinal 14-3-3β expression ameliorated the developed allodynia. Moreover, accompanied with allodynia, SNL provoked HDAC4 phosphorylation–dependent HDAC4-14-3-3β coupling that restricted location of HDAC4 to the cytoplasm. Although it failed to affect HDAC4 phosphorylation, knockdown of spinal 14-3-3β expression uncoupled HDAC4 from 14-3-3, attenuated cytoplasmic HDAC4 retention, and ameliorated the developed allodynia. In line with evidences showing phosphorylation-dependent HDAC4 coupling with 14-3-3β induces cytosolic HDAC4 accumulation7  regulates memory-associated synaptic plasticity.30–32  Our results imply that HDAC4-14-3-3β coupling-dependent cytoplasmic HDAC4 retention is crucial for the neuropathic development, and this machinery is possibly involved in activity-dependent plasticity in general.

To understand the machinery underlying the sensitization of nociception may eventually provide insight into developing medical strategies for pain relief. In this study, we identified spinal HDAC4 functions as a key element important for the neuropathic injury–associated allodynia. Although the use of HDACs-targeting siRNAs/antagonists for therapy garners lots of interest from researchers, we focally knocked-down 14-3-3β instead of HDAC4 expression. Our choice is based on that HDAC4 is abundantly expressed in the central nervous system of mammalian,33  and selective HDAC4 knockout, even restricted to the central nervous system, could abolish its inhibition of gene transcription required for normal neural activity, despite it might ameliorate pain hypersensitivity. Our speculation is supported by mounting evidence linking misregulated HDAC activity to degenerative disease models such as Huntington disease,34  Parkinson disease,35  Alzheimer disease,36  motor neuron disorders,37  and Rubinstein–Taybi syndrome.38  In contrast, our results showed trim-down of spinal 14-3-3β exhibited almost no effect on the neuropathic injury–induced HDAC4 phosphorylation, implying this protocol has minor impact on HDAC4 activity. We, therefore, suggest that designing 14-3-3β-targeting siRNAs/drugs would be a good starting point for developing medical treatments for neuropathic pain. However, possible side effects of candidate agents need to be verified carefully.

This research was supported by the National Science Council of Taiwan (Taipei, Taiwan, Republic of China; grant nos. NSC 102-2628-B-715-001 and 101-2320-B-715-001-MY3 to Dr. Peng, and NSC 101-2320-B-039-013-MY3 to Dr. Lin) and by the Mackay Medical College (New Taipei, Taiwan, Republic of China; grant nos. MMH-MM-10206 and MMH-MM-10302 to Dr. Peng) and Taipei Medical University (Taipei, Taiwan, Republic of China; grant no. TMU102-AE1-B06 to Dr. Lin).

The authors declare no competing interests.

1.
Woolf
CJ
:
Central sensitization: Implications for the diagnosis and treatment of pain.
Pain
2011
;
152
(
3 suppl
):
S2
15
2.
Bai
G
,
Wei
D
,
Zou
S
,
Ren
K
,
Dubner
R
:
Inhibition of class II histone deacetylases in the spinal cord attenuates inflammatory hyperalgesia.
Mol Pain
2010
;
6
:
51
3.
Sailaja
BS
,
Cohen-Carmon
D
,
Zimmerman
G
,
Soreq
H
,
Meshorer
E
:
Stress-induced epigenetic transcriptional memory of acetylcholinesterase by HDAC4.
Proc Natl Acad Sci U S A
2012
;
109
:
E3687
95
4.
Rajan
I
,
Savelieva
KV
,
Ye
GL
,
Wang
CY
,
Malbari
MM
,
Friddle
C
,
Lanthorn
TH
,
Zhang
W
:
Loss of the putative catalytic domain of HDAC4 leads to reduced thermal nociception and seizures while allowing normal bone development.
PLoS One
2009
;
4
:
e6612
5.
Haberland
M
,
Montgomery
RL
,
Olson
EN
:
The many roles of histone deacetylases in development and physiology: Implications for disease and therapy.
Nat Rev Genet
2009
;
10
:
32
42
6.
McKinsey
TA
,
Zhang
CL
,
Olson
EN
:
Identification of a signal-responsive nuclear export sequence in class II histone deacetylases.
Mol Cell Biol
2001
;
21
:
6312
21
7.
Grozinger
CM
,
Schreiber
SL
:
Regulation of histone deacetylase 4 and 5 and transcriptional activity by 14-3-3-dependent cellular localization.
Proc Natl Acad Sci U S A
2000
;
97
:
7835
40
8.
Gannon-Murakami
L
,
Murakami
K
:
Selective association of protein kinase C with 14-3-3ζ in neuronally differentiated PC12 Cells. Stimulatory and inhibitory effect of 14-3-3ζ in vivo.
J Biol Chem
2002
;
277
:
23116
22
9.
Chandran
S
,
Li
H
,
Dong
W
,
Krasinska
K
,
Adams
C
,
Alexandrova
L
,
Chien
A
,
Hallows
KR
,
Bhalla
V
:
Neural precursor cell-expressed developmentally down-regulated protein 4-2 (Nedd4-2) regulation by 14-3-3 protein binding at canonical serum and glucocorticoid kinase 1 (SGK1) phosphorylation sites.
J Biol Chem
2011
;
286
:
37830
40
10.
Laffray
S
,
Bouali-Benazzouz
R
,
Papon
MA
,
Favereaux
A
,
Jiang
Y
,
Holm
T
,
Spriet
C
,
Desbarats
P
,
Fossat
P
,
Le Feuvre
Y
,
Decossas
M
,
Héliot
L
,
Langel
U
,
Nagy
F
,
Landry
M
:
Impairment of GABAB receptor dimer by endogenous 14-3-3ζ in chronic pain conditions.
EMBO J
2012
;
31
:
3239
51
11.
Walkinshaw
DR
,
Weist
R
,
Kim
GW
,
You
L
,
Xiao
L
,
Nie
J
,
Li
CS
,
Zhao
S
,
Xu
M
,
Yang
XJ
:
The tumor suppressor kinase LKB1 activates the downstream kinases SIK2 and SIK3 to stimulate nuclear export of class IIa histone deacetylases.
J Biol Chem
2013
;
288
:
9345
62
12.
Kobayashi
T
,
Deak
M
,
Morrice
N
,
Cohen
P
:
Characterization of the structure and regulation of two novel isoforms of serum- and glucocorticoid-induced protein kinase.
Biochem J
1999
;
344
(
Pt 1
):
189
97
13.
Peng
HY
,
Chen
GD
,
Hsieh
MC
,
Lai
CY
,
Huang
YP
,
Lin
TB
:
Spinal SGK1/GRASP-1/Rab4 is involved in complete Freund’s adjuvant-induced inflammatory pain via regulating dorsal horn GluR1-containing AMPA receptor trafficking in rats.
Pain
2012
;
153
:
2380
92
14.
Peng
HY
,
Chen
GD
,
Lai
CY
,
Hsieh
MC
,
Lin
TB
:
Spinal serum-inducible and glucocorticoid-inducible kinase 1 mediates neuropathic pain via kalirin and downstream PSD-95-dependent NR2B phosphorylation in rats.
J Neurosci
2013
;
33
:
5227
40
15.
Zimmermann
M
:
Ethical guidelines for investigations of experimental pain in conscious animals.
Pain
1983
;
16
:
109
10
16.
Kim
SH
,
Chung
JM
:
An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat.
Pain
1992
;
50
:
355
63
17.
Peng
HY
,
Chen
GD
,
Lee
SD
,
Lai
CY
,
Chiu
CH
,
Cheng
CL
,
Chang
YS
,
Hsieh
MC
,
Tung
KC
,
Lin
TB
:
Neuroactive steroids inhibit spinal reflex potentiation by selectively enhancing specific spinal GABAA receptor subtypes.
Pain
2009
;
143
:
12
20
18.
Peng
HY
,
Chang
CH
,
Tsai
SJ
,
Lai
CY
,
Tung
KC
,
Wu
HC
,
Lin
TB
:
Protein kinase A-dependent spinal α-amino-3-hydroxy-5-methyl-4-isoxazoleproprionate-receptor trafficking mediates capsaicin-induced colon-urethra cross-organ reflex sensitization.
Anesthesiology
2011
;
114
:
70
83
19.
Schäfers
M
,
Svensson
CI
,
Sommer
C
,
Sorkin
LS
:
Tumor necrosis factor-α induces mechanical allodynia after spinal nerve ligation by activation of p38 MAPK in primary sensory neurons.
J Neurosci
2003
;
23
:
2517
21
20.
Hori
K
,
Ozaki
N
,
Suzuki
S
,
Sugiura
Y
:
Upregulations of P2X(3) and ASIC3 involve in hyperalgesia induced by cisplatin administration in rats.
Pain
2010
;
149
:
393
405
21.
Bottomley
MJ
,
Lo Surdo
P
,
Di Giovine
P
,
Cirillo
A
,
Scarpelli
R
,
Ferrigno
F
,
Jones
P
,
Neddermann
P
,
De Francesco
R
,
Steinkühler
C
,
Gallinari
P
,
Carfí
A
:
Structural and functional analysis of the human HDAC4 catalytic domain reveals a regulatory structural zinc-binding domain.
J Biol Chem
2008
;
283
:
26694
704
22.
Park
H
,
Kim
S
,
Kim
YE
,
Lim
SJ
:
A structure-based virtual screening approach toward the discovery of histone deacetylase inhibitors: Identification of promising zinc-chelating groups.
ChemMedChem
2010
;
5
:
591
7
23.
Sinnett-Smith
J
,
Ni
Y
,
Wang
J
,
Ming
M
,
Young
SH
,
Rozengurt
E
:
Protein kinase D1 mediates class IIa histone deacetylase phosphorylation and nuclear extrusion in intestinal epithelial cells: Role in mitogenic signaling.
Am J Physiol Cell Physiol
2014
;
306
:
C961
71
24.
Backs
J
,
Song
K
,
Bezprozvannaya
S
,
Chang
S
,
Olson
EN
:
CaM kinase II selectively signals to histone deacetylase 4 during cardiomyocyte hypertrophy.
J Clin Invest
2006
;
116
:
1853
64
25.
Aitken
A
:
14-3-3 and its possible role in co-ordinating multiple signalling pathways.
Trends Cell Biol
1996
;
6
:
341
7
26.
Pawson
T
,
Scott
JD
:
Signaling through scaffold, anchoring, and adaptor proteins.
Science
1997
;
278
:
2075
80
27.
Wang
AH
,
Kruhlak
MJ
,
Wu
J
,
Bertos
NR
,
Vezmar
M
,
Posner
BI
,
Bazett-Jones
DP
,
Yang
XJ
:
Regulation of histone deacetylase 4 by binding of 14-3-3 proteins.
Mol Cell Biol
2000
;
20
:
6904
12
28.
Reiter
MH
,
Vila
G
,
Knosp
E
,
Baumgartner-Parzer
SM
,
Wagner
L
,
Stalla
GK
,
Luger
A
:
Opposite effects of serum- and glucocorticoid-regulated kinase-1 and glucocorticoids on POMC transcription and ACTH release.
Am J Physiol Endocrinol Metab
2011
;
301
:
E336
41
29.
Martin
H
,
Rostas
J
,
Patel
Y
,
Aitken
A
:
Subcellular localisation of 14-3-3 isoforms in rat brain using specific antibodies.
J Neurochem
1994
;
63
:
2259
65
30.
Qiao
H
,
Foote
M
,
Graham
K
,
Wu
Y
,
Zhou
Y
:
14-3-3 proteins are required for hippocampal long-term potentiation and associative learning and memory.
J Neurosci
2014
;
34
:
4801
8
31.
Skoulakis
EM
,
Davis
RL
:
14-3-3 proteins in neuronal development and function.
Mol Neurobiol
1998
;
16
:
269
84
32.
Sando
R
III
,
Gounko
N
,
Pieraut
S
,
Liao
L
,
Yates
J
III
,
Maximov
A
:
HDAC4 governs a transcriptional program essential for synaptic plasticity and memory.
Cell
2012
;
151
:
821
34
33.
Kim
MS
,
Akhtar
MW
,
Adachi
M
,
Mahgoub
M
,
Bassel-Duby
R
,
Kavalali
ET
,
Olson
EN
,
Monteggia
LM
:
An essential role for histone deacetylase 4 in synaptic plasticity and memory formation.
J Neurosci
2012
;
32
:
10879
86
34.
Hockly
E
,
Richon
VM
,
Woodman
B
,
Smith
DL
,
Zhou
X
,
Rosa
E
,
Sathasivam
K
,
Ghazi-Noori
S
,
Mahal
A
,
Lowden
PA
,
Steffan
JS
,
Marsh
JL
,
Thompson
LM
,
Lewis
CM
,
Marks
PA
,
Bates
GP
:
Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington’s disease.
Proc Natl Acad Sci U S A
2003
;
100
:
2041
6
35.
Gardian
G
,
Yang
L
,
Cleren
C
,
Calingasan
NY
,
Klivenyi
P
,
Beal
MF
:
Neuroprotective effects of phenylbutyrate against MPTP neurotoxicity.
Neuromolecular Med
2004
;
5
:
235
41
36.
Ricobaraza
A
,
Cuadrado-Tejedor
M
,
Pérez-Mediavilla
A
,
Frechilla
D
,
Del Río
J
,
García-Osta
A
:
Phenylbutyrate ameliorates cognitive deficit and reduces tau pathology in an Alzheimer’s disease mouse model.
Neuropsychopharmacology
2009
;
34
:
1721
32
37.
Echaniz-Laguna
A
,
Bousiges
O
,
Loeffler
JP
,
Boutillier
AL
:
Histone deacetylase inhibitors: Therapeutic agents and research tools for deciphering motor neuron diseases.
Curr Med Chem
2008
;
15
:
1263
73
38.
Alarcón
JM
,
Malleret
G
,
Touzani
K
,
Vronskaya
S
,
Ishii
S
,
Kandel
ER
,
Barco
A
:
Chromatin acetylation, memory, and LTP are impaired in CBP+/− mice: A model for the cognitive deficit in Rubinstein-Taybi syndrome and its amelioration.
Neuron
2004
;
42
:
947
59