Background

Liver X receptors, including α and β isoforms, are ligand-activated transcription factors. Whether liver X receptor α plays a role in neuropathic pain is unknown.

Methods

A spared nerve injury model was established in adult male rats and mice. Von Frey tests were performed to evaluate the neuropathic pain behavior; Western blot and immunohistochemistry were performed to understand the underlying mechanisms.

Results

Intrathecal injection of a specific liver X receptor agonist T0901317 or GW3965 could either prevent the development of mechanical allodynia or alleviate the established mechanical allodynia, both in rats and wild-type mice. GW3965 could inhibit the activation of glial cells and the expression of tumor necrosis factor-α (mean ± SD: 196 ± 48 vs. 119 ± 57; n = 6; P < 0.01) and interleukin 1β (mean ± SD: 215 ± 69 vs. 158 ± 74; n = 6; P < 0.01) and increase the expression of interleukin 10 in the spinal dorsal horn. All of the above effects of GW3965 could be abolished by liver X receptor α mutation. Moreover, more glial cells were activated, and more interleukin 1β was released in the spinal dorsal horn in liver X receptor α knockout mice than in wild-type mice after spared nerve injury. Aminoglutethimide, a neurosteroid synthesis inhibitor, blocked the inhibitory effect of T0901317 on mechanical allodynia, on the activation of glial cells, and on the expression of cytokines.

Conclusions

Activation of liver X receptor α inhibits mechanical allodynia by inhibiting the activation of glial cells and rebalancing cytokines in the spinal dorsal horn via neurosteroids.

What We Already Know about This Topic
  • Ligands for liver X receptors (LXRs, which are nominally intracellular cholesterol sensors), can decrease lipopolysaccharide-induced expression of proinflammatory genes in activated macrophages

  • An LXR ligand can reduce neuroinflammation after spinal cord injury, but effects on neuropathic pain are not known

What This Article Tells Us That Is New
  • In male rodent models of spared nerve injury, intrathecal liver X receptor (LXR) agonists reduced mechanical allodynia

  • This effect was not observed in animals with a mutation in the LXRα receptor subtype

  • LXR agonist inhibited glial cell activation and expression of cytokines in the spinal dorsal horn

NEUROPATHIC pain is a debilitating pain state, often caused by lesion or disease of the peripheral or central nervous system.1  The management of neuropathic pain is still challenging, because it does not respond to most currently used analgesic drugs.

Liver X receptors (LXRs) are considered as intracellular cholesterol sensors, for which the activation leads to decreased plasma cholesterol.2  Two known different isoforms, LXRα and LXRβ, have been recognized. LXRα is mainly expressed in the liver, whereas LXRβ is broadly expressed.3,4  LXRs ligands, including oxysterols GW3965 and T0901317, decrease lipopolysaccharide-induced expression of proinflammatory genes in activated macrophages.5  T0901317 has recently been used to reduce neuroinflammation after spinal cord injury.6  However, whether LXRα plays a role in neuropathic pain is still unknown.

In response to nervous tissue damage, spinal microglia and astrocytes were activated, resulting in overproduction of inflammatory cytokines, such as tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), interleukin 6, and thus contributing to neuropathic pain.7–11  In contrast to proinflammatory cytokines, the antiinflammatory cytokine interleukin 10 (IL-10) has been shown to be gradually reduced as neuropathic pain develops.12  Intrathecal injection of plasmid DNA encoding IL-10 or recombinant rat IL-10 could suppress neuropathic pain symptoms.13,14 

Therefore, we hypothesized that activation of LXRα could inhibit the neuroinflammatory responses in the spinal cord and thereby alleviate neuropathic pain. Here we first studied the time course of LXRα expression in the lumbar spinal dorsal horn after spared nerve injury and then investigated the effects of LXR agonists on the development and maintenance of mechanical allodynia. The mechanisms by which LXR agonists inhibit mechanical allodynia were also explored.

Animals

Male Sprague–Dawley rats weighing 180 to 200 g and LXRα knockout (LXRα [–/–]) C57 and wild-type male mice weighing 25 to 31g were used in this study. LXRα (–/–) mice were purchased from Jackson Laboratory (USA). Animals were group housed, with free access to standard rodent chow and water. The animals were exposed to a 12-h light/dark cycle. All of the studies were approved by the Sun Yat-Sen University Animal Care and Use Committee (Guangzhou, Guangdong, People’s Republic of China) and were carried out in accordance with the guidelines of the National Institutes of Health on animal care and the ethical guidelines for investigation of experimental pain in conscious animals.15  All of the animals were randomly assigned to different treatment groups.

In total, 424 rats and 171 mice were used in the present study; among them, three rats and one mouse died after surgery, two rats were euthanized due to excessive suffering from pain. Thirty-five rats and 17 mice were excluded because no mechanical allodynia was induced after spared nerve injury surgery or because catheterization was unsuccessful.

Drugs Administration

Two synthetic LXR agonists, T0901317 and GW3965, were used in the current study. The EC50 of T0901317, which activates both LXRα and LXRβ, is approximately 20 nM.16  GW3965 has a greater affinity toward LXRβ (EC50 = 30 nM) than LXRα (EC50 = 190 nM).17  T0901317 was purchased from Cayman Chemical (USA). GW3965 and aminoglutethimide (R(+)-p-aminoglutethimide) were purchased from Sigma-Aldrich (USA). All of the drugs were dissolved in dimethyl sulfoxide. T0901317 and GW3965 were injected intrathecally with polyethylene 10, for which the tip was put on the spinal lumbar enlargement level 1 week before. Whether polyethylene 10 was correctly placed in intervertebral space was confirmed by paralysis in the bilateral hind limb after 2% lidocaine injection (7 µl for rats and 3 µl for mice) through the tube within 30 s. Drugs or vehicle was administered slowly in volumes of 10 µl (for rats) or 3 µl (for mice).

Neuropathic Pain Model: Spared Nerve Injury

Spared nerve injury was carried out following the previous method.18  Briefly, rats and mice were anesthetized with isoflurane (1.5 to 2.5%) and a mixture of 30% N2O and 70% O2. The sciatic nerve of the left leg was exposed into common peroneal, tibial nerve, and sural nerve, and then the common peroneal and tibial nerves were ligated and cut, whereas the sural nerve was left intact. For the animals in the sham group, the sciatic nerve was only exposed but not ligated or cut.

Assessment of Mechanical Sensitivity

Mechanical sensitivity of the animals before and after spared nerve injury was assessed with the up–down method following the previously described method.19  Briefly, after habituation for 10 to 15 min (rats) or 45 min to 1 h (mice), a series of filaments with varying forces (0.4, 0.6, 1, 1.4, 2, 4, 6, 8, and 15 g for rats and 0.04, 0.07, 0.16, 0.4, 0.6, 1, 1.4, and 2 g for mice) were applied to the plantar surface of the hind paw. Each stimulus consisted of a maximum 6-s application of the filament, and quick withdrawal in response to the stimulus was considered a positive response. The 50% paw withdrawal thresholds were then calculated.

To test whether T0901317 could affect the mechanical sensitivity in the sham-operated rats, the incidence of paw withdrawal to four different forces of filaments (2, 6, 15, and 26 g) was measured according to previous work.20  Each filament was applied once per second to the plantar surface eight times. Ten trials were performed on each hind paw. The percentage of positive trials was recorded. The behavioral test was performed in a blinded fashion.

Western Blotting

Animals were anesthetized with sodium pentobarbital (50 mg/kg) after defined survival time. If the animals received T0901317 or GW3965 treatment, the tissues were collected 2 days after injection. The ipsilateral spinal dorsal horn was dissected and homogenized in 15 mM Tris buffer (pH 7.6; 250 mM sucrose, 1 mM magnesium chloride, 1 mM dithiothreitol, 2.5 mM EDTA, 1 mM EGTA, 50 mM sodium fluoride, 10 μg/ml leupeptin, 1.25 μg/ml pepstatin, 2.5 μg/ml aprotin, 2 mM sodium pyrophosphate, 0.1 mM sodium orthovanadate, 0.5 mM phenylmethylsulfonyl fluoride, and protease inhibitor mixture; Roche Molecular Biochemicals, Switzerland). The samples were sonicated and then centrifuged at 13,000g for 15 min. The isolated proteins were separated by sodium dodecyl sulphate–polyacrylamide gel electrophoresis and transferred onto a polyvinylidene fluoride membrane (Bio-Rad Laboratories, Inc., USA). After incubation with blocking buffer for 1 h at room temperature, the blots were incubated with primary antibody against IL-1β (1:200, Abcam; United Kingdom), IL-10 antibody (1:500, Abcam), or TNFα (1:200; Santa Cruz Biotechnology, Inc., USA) overnight at 4°C, followed by incubation with secondary antibody horseradish peroxidase–conjugated rabbit antigoat, goat antimouse, or goat antirabbit immunoglobulin G (1:8000, 1:5000, or 1:8000; Kirkegaard & Perry Laboratories, Inc., USA) for 2 h at room temperature. The blots were developed with enhanced chemiluminescence (Clarity Western electrochemiluminescence substrate, Bio-Rad Laboratories, Inc.) and detected by a Tanon 5200 imager (Tanon Science & Technology Co., Ltd., China). The intensities of the blots were quantified by Tanon MP (Tanon Science & Technology Co., Ltd., China) software and normalized against a loading control (β actin).

Immunohistochemistry

Immunohistochemistry was performed following our previous study.21  Briefly, rats or mice were randomly chosen from a cohort used for behavioral studies. If the animals received T0901317 or GW3965 treatment, the tissues were collected 2 days after injection. After defined survival times, the animals were anesthetized and perfused with saline followed by 4% paraformaldehyde in 0.1 M phosphate buffer. The lumbar spinal cord segments were removed and postfixed in the 4% paraformaldehyde for 3 h and then replaced with 30% sucrose overnight. Cryostat sections (25 μm) were cut in a cryostat (Leica CM1900, Leica Biosystems, Germany) and processed for immunohistochemistry. For double immunofluorescence staining, primary antibody for LXRα (1:200; Abcam) was incubated together with antibody for mouse monoclonal neuronal-specific nuclear protein (neuronal marker, 1:200; Millipore Bioscience Research Reagents, USA), glial fibrillary acidic protein ([GFAP] astrocyte marker, 1:1000; Cell Signaling Technology, USA), or goat polyclonal antiionized calcium-binding adaptor molecule 1 ([Iba1] microglia marker, 1:800; Abcam). After incubation for more than two nights at 4°C, the sections were incubated with cy3-conjugated (1:500; Jackson Immuno-Research, USA) and fluorescein isothiocyanate–conjugated secondary antibodies (1:400; Jackson ImmunoResearch) for 1 h at room temperature. For negative control sections, the above procedures were followed, except the primary antibody was omitted. The stained sections were then examined with a Leica fluorescence microscope (Leica Biosystems), and images were captured with a Leica DFC350 FX camera (Leica Camera AG). For quantification of immunofluorescence staining, the area of LXRα- immunoreactivity per section was measured in the spinal dorsal horn (laminae I to V) using a Leica Qwin V3 digital-image processing system (Leica Camera AG).

Quantification and Statistics

Data were expressed as mean ± SD and analyzed with SPSS 15.0 (SPSS Inc., USA). The data of behavioral tests were analyzed using repeated-measures two-way ANOVA with the Tukey post hoc test. Immunohistochemistry and Western blot data were analyzed by one-way ANOVA followed by the Tukey post hoc test. P < 0.05 was considered statistically significant.

LXRα Was Up-regulated in the Lumbar Spinal Dorsal Horn and Lumbar 5 Dorsal Root Ganglia After Spared Nerve Injury

Compared with the sham group (fig. 1A, reference value of 100), the expression of LXRα in the ipsilateral (fig. 1A) and contralateral (fig. 1B) lumbar spinal dorsal horns increased significantly from day 1 to day 14 after the surgery. Immunohistochemistry showed that, compared with the sham group (fig. 1C), the expression of LXRα was increased 1 (fig. 1D) and 7 days (fig. 1E) after spared nerve injury. LXRα was increased in the bilateral spinal dorsal horn 7 days after spared nerve injury (fig. 1F). In the negative control group (fig. 1G), no signals were detected. Compared with the sham group, the areas of LXRα immunoreactivity were all increased in laminae I to II, III to IV, and V in the ipsilateral spinal dorsal horn (fig. 1H).

Fig. 1.

Liver X receptor (LXR) α was up-regulated in the spinal dorsal horn after spared nerve injury. Western blot shows the expression of LXRα in the ipsilateral (A) and contralateral (B) lumbar spinal dorsal horn 1, 4, 7, 14, and 21 days after spared nerve injury (SNI) and 7 days after sham operation. The quantification of LXRα normalized by β-actin is shown under the representative bands (n = 5/group). (C through E) Changes of LXRα infrared (IR) in the ipsilateral lumbar spinal dorsal horn from sham-operated and SNI rats. (F) Changes of LXRα IR in bilateral lumbar spinal dorsal horn 7 days after SNI surgery. (G) Representative experiments show the results of negative controls (n = 5). (H) Quantification of LXRα IR–positive area in different lamina in ipsilateral lumbar spinal dorsal horn 7 days after sham or SNI surgery (n = 5/group). *P < 0.05, **P < 0.01, ***P < 0.001 versus sham group. Scale bars: in C through F, 100 µm; in G, 200 µm.

Fig. 1.

Liver X receptor (LXR) α was up-regulated in the spinal dorsal horn after spared nerve injury. Western blot shows the expression of LXRα in the ipsilateral (A) and contralateral (B) lumbar spinal dorsal horn 1, 4, 7, 14, and 21 days after spared nerve injury (SNI) and 7 days after sham operation. The quantification of LXRα normalized by β-actin is shown under the representative bands (n = 5/group). (C through E) Changes of LXRα infrared (IR) in the ipsilateral lumbar spinal dorsal horn from sham-operated and SNI rats. (F) Changes of LXRα IR in bilateral lumbar spinal dorsal horn 7 days after SNI surgery. (G) Representative experiments show the results of negative controls (n = 5). (H) Quantification of LXRα IR–positive area in different lamina in ipsilateral lumbar spinal dorsal horn 7 days after sham or SNI surgery (n = 5/group). *P < 0.05, **P < 0.01, ***P < 0.001 versus sham group. Scale bars: in C through F, 100 µm; in G, 200 µm.

Close modal

In the gray matter, LXRα was located mainly in neurons (fig. 2A) but not in astrocytes (fig. 2B) or microglia (fig. 2C) 7 days after sham operation, and 7 days after spared nerve injury, LXRα was still located mainly in neurons (fig. 2D) but not in astrocytes (fig. 2E) or microglia (fig. 2F). In the white matter, LXRα was located mainly in neurite outgrowth inhibitor A–labeled oligodendrocytes (fig. 2G) but not in astrocytes (fig. 2H) and microglia (fig. 2I) in sham-operated rats, whereas 7 days after spared nerve injury, LXRα was located mainly in oligodendrocytes (fig. 2J) and to a lesser extent in astrocytes (fig. 2K) and microglia (fig. 2L).

Fig. 2.

The cell types that express liver X receptor (LXR) α in spinal dorsal horn from sham and spared nerve injury (SNI) rats. (A through F) In gray matter of the ipsilateral lumbar spinal dorsal horn, LXRα was located mainly in neuronal-specific nuclear protein (NeuN) –labeled neurons (A and D) but not in glial fibrillary acidic protein (GFAP) –labeled astrocyte (B and E) or in ionized calcium binding adaptor molecule 1 (Iba1) –labeled microglia (C and F) 7 days after sham and SNI operation. (G through L) In the white matter, LXRα was located mainly in oligodendrocytes (G) but not in astrocytes (H) and microglia (I) 7 days after sham operation. Seven days after SNI, LXRα was still located mainly in oligodendrocytes (J) but to a lesser extent in astrocytes (K) and microglia (L). Bar = 50 µm. Nogo-A = neurite outgrowth inhibitor A.

Fig. 2.

The cell types that express liver X receptor (LXR) α in spinal dorsal horn from sham and spared nerve injury (SNI) rats. (A through F) In gray matter of the ipsilateral lumbar spinal dorsal horn, LXRα was located mainly in neuronal-specific nuclear protein (NeuN) –labeled neurons (A and D) but not in glial fibrillary acidic protein (GFAP) –labeled astrocyte (B and E) or in ionized calcium binding adaptor molecule 1 (Iba1) –labeled microglia (C and F) 7 days after sham and SNI operation. (G through L) In the white matter, LXRα was located mainly in oligodendrocytes (G) but not in astrocytes (H) and microglia (I) 7 days after sham operation. Seven days after SNI, LXRα was still located mainly in oligodendrocytes (J) but to a lesser extent in astrocytes (K) and microglia (L). Bar = 50 µm. Nogo-A = neurite outgrowth inhibitor A.

Close modal

We found that LXRα was also up-regulated in lumbar 5 dorsal root ganglia after spared nerve injury (Supplemental Digital Content 1A, http://links.lww.com/ALN/B469). LXRα was expressed in neurofilament protein 200 and isolectin B4–labeled neurons but not in GFAP-labeled satellite cells (Supplemental Digital Content 1B, http://links.lww.com/ALN/B469).

Intrathecal Injection of LXRs Agonist Either Before or After Spared Nerve Injury Attenuated Mechanical Allodynia Induced by Spared Nerve Injury

Additional experiments were performed to test whether the activation of LXRs could affect the development and maintenance of mechanical allodynia. We found that intrathecal injection of T0901317, started 30 min before spared nerve injury, and thereafter once daily for 2 days, blocked mechanical allodynia dose dependently. T0901317 at 19.2 µg completely blocked the decrease in the bilateral 50% paw withdrawal threshold on the ipsilateral hind paw, and the effect persisted until the end of the experiment. T0901317 at 4.8 µg produced a weaker antiallodynia effect (fig. 3A). In contrast, in the dimethyl sulfoxide–treated rats, 50% paw withdrawal threshold decreased in the bilateral hind paw 4 days after spared nerve injury, which persisted for at least 21 days (fig. 3, A and B). A single intrathecal injection of T0901317 (19.2 µg) 7 days after spared nerve injury reversed the decrease of the 50% paw withdrawal threshold in the bilateral side (fig. 3C). T0901317 could induce expression not only of LXR target genes but also of the nuclear xenobiotic receptor pregnane X receptor target genes, a characteristic that more specific LXR agonists such as GW3965 do not have.17,22  In the present study, to test whether the effect of T0901317 was specific, GW3965 was also injected intrathecally into spared nerve injury rats. As shown in figure 3D, GW3965 (6.2 µg) could also attenuate the mechanical allodynia when applied 7 days after spared nerve injury. A 50% paw withdrawal threshold was also increased for 1 day when 19.2 µg T0901317 was administrated 21 days after spared nerve injury (fig. 3E). For the paw withdrawal incidence, no significant difference between the dimethyl sulfoxide- and T0901317-treated groups (19.2 µg) was found (fig. 3F).

Fig. 3.

Intrathecal (IT) injection of liver X receptor (LXR) α/β agonist either before or after spared nerve injury (SNI) attenuated mechanical allodynia induced by SNI. (A, B) T0901317 dose-dependently blocked bilateral mechanical allodynia induced by SNI (n = 5 or 6/group). (C, D) A single IT injection of T0901317 (19.2 µg [C]; n = 7) or GW3965 (6.2 µg [D]; n = 7) 7 days after SNI increased the 50% paw withdrawal threshold in the bilateral side (n = 6/group). (E) A single IT injection of T0901317 into rats that had received SNI surgery 21 days before inhibition of the mechanical allodynia 1 and 2 h after injection. (F) In the sham-operated rats, T0901317 at 19.2 µg had no effect on the paw withdrawal incidence evoked by the four different filaments (n = 5). Arrows show the injection of T0901317 or GW3965. *P < 0.05, **P < 0.01, ***P < 0.001 versus baseline; $$P < 0.01, $$$P < 0.001 versus dimethyl sulfoxide–treated SNI group. DMSO = dimethyl sulfoxide.

Fig. 3.

Intrathecal (IT) injection of liver X receptor (LXR) α/β agonist either before or after spared nerve injury (SNI) attenuated mechanical allodynia induced by SNI. (A, B) T0901317 dose-dependently blocked bilateral mechanical allodynia induced by SNI (n = 5 or 6/group). (C, D) A single IT injection of T0901317 (19.2 µg [C]; n = 7) or GW3965 (6.2 µg [D]; n = 7) 7 days after SNI increased the 50% paw withdrawal threshold in the bilateral side (n = 6/group). (E) A single IT injection of T0901317 into rats that had received SNI surgery 21 days before inhibition of the mechanical allodynia 1 and 2 h after injection. (F) In the sham-operated rats, T0901317 at 19.2 µg had no effect on the paw withdrawal incidence evoked by the four different filaments (n = 5). Arrows show the injection of T0901317 or GW3965. *P < 0.05, **P < 0.01, ***P < 0.001 versus baseline; $$P < 0.01, $$$P < 0.001 versus dimethyl sulfoxide–treated SNI group. DMSO = dimethyl sulfoxide.

Close modal

Effects of LXR Agonists on Mechanical Allodynia Were Abolished in LXRα (–/–) Mice

Intrathecal injection of T0901317 (4.4 µg) significantly increased the bilateral 50% paw withdrawal threshold in wild-type mice 1 to 3 days after the injection, whereas the same dose of T0901317 had no effect on the decrease of 50% paw withdrawal threshold in these LXRα (–/–) mice at the same time points (fig. 4, A and B). The analgesic effect of GW3965 (5.6 µg) was also completely abolished in LXRα (–/–) mice (fig. 4, C and D). Activation of astrocyte and microglia in the spinal cord was enhanced in LXRα (–/–) mice after spared nerve injury, and GW3965 inhibited activation of glial cells, decreased the expression of IL-1β and TNF-α, and increased the expression of IL-10 in the spinal dorsal horn induced by spared nerve injury in wild-type but not in LXRα (–/–) mice.

Fig. 4.

The effects of liver X receptor (LXR) agonists on mechanical allodynia induced by spared nerve injury (SNI) was abolished in LXRα knockout ([–/–]) mice. (A, B) Intrathecal injection of T0901317 (4.4 µg) significantly increased bilateral 50% paw withdrawal threshold in wild-type (n = 11) but not LXRα ([–/–] n = 10) mice 1, 4, and 7 days after the drug injection. (C, D) Compared with wild-type mice (n = 13), the inhibitory effect of GW3965 (5.6 µg) was also completely abolished in LXRα (–/–) mice (n = 14). *P < 0.05, **P < 0.01, ***P < 0.001 versus LXRα (–/–) mice that received SNI surgery. Arrows indicate the time of T0901317 or GW3965 injection.

Fig. 4.

The effects of liver X receptor (LXR) agonists on mechanical allodynia induced by spared nerve injury (SNI) was abolished in LXRα knockout ([–/–]) mice. (A, B) Intrathecal injection of T0901317 (4.4 µg) significantly increased bilateral 50% paw withdrawal threshold in wild-type (n = 11) but not LXRα ([–/–] n = 10) mice 1, 4, and 7 days after the drug injection. (C, D) Compared with wild-type mice (n = 13), the inhibitory effect of GW3965 (5.6 µg) was also completely abolished in LXRα (–/–) mice (n = 14). *P < 0.05, **P < 0.01, ***P < 0.001 versus LXRα (–/–) mice that received SNI surgery. Arrows indicate the time of T0901317 or GW3965 injection.

Close modal

We further investigated whether LXRα modulated astrocytes and microglial activation (fig. 5). We found that, compared with the wild-type mice (fig. 5, Aa and Ag), 9 days after spared nerve injury, astrocytes (fig. 5Ac) and microglia (fig. 5Ai) in the ipsilateral lumbar spinal dorsal horn were highly activated, because the expression of GFAP and Iba1 was increased and the size of the cells was enlarged. Knockout of LXRα alone did not induce activation of astrocyte (fig. 5Ab) and microglia (fig. 5Ah); however, knockout of LXRα activated more astrocytes in the spinal dorsal horn 9 days after spared nerve injury, as shown in figure 5Ad and figure 5Aj. GW3965 could only inhibit the activation of astrocytes (fig. 5Ae) and microglia (fig. 5Ak) in the wild-type mice but not in the LXRα (–/–) mice, because we can see that the shapes and numbers of astrocytes and microglia in the GW3965-treated (LXRα [–/–]) group were comparable with those in the dimethyl sulfoxide–treated LXRα (–/–) group (fig. 5, Af and Al). Furthermore, treatment of GW3965 could decrease the expression of IL-1β (fig. 5D; 158 ± 74) and TNF-α (fig. 5E; 119 ± 57) and increase the expression of IL-10 (fig. 5F) in the wild-type mice that received spared nerve injury (215 ± 69 for IL-1β; 196 ± 48 for TNF-α; and 63 ± 22 for IL-10) but not in the LXRα (–/–) mice that received spared nerve injury, 9 days after spared nerve injury, the expression of IL-1β, but not IL-10 and TNF-α was increased in LXRα (–/–) mice, compared to the wild-type mice.

Fig. 5.

Activation of astrocyte and microglia in the spinal cord was enhanced in liver X receptor (LXR) α knockout ([–/–]) mice after spared nerve injury (SNI), and GW3965 inhibited activation of astrocytes and microglia, down-regulated interleukin (IL) 1β and tumor necrosis factor (TNF) α, and up-regulated IL-10 expression in the spinal dorsal horn in wild-type but not in LXRα (–/–) mice. (A) Expression of glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1) in six different groups is shown. (B, C) The histogram shows the summary data of the GFAP- or Iba1-positive area in different groups (n = 6/group). (D through F) Protein levels of IL-1β, TNF-α, IL-10, and β-actin from samples of ipsilateral spinal dorsal horn in eight groups. The histogram shows the quantification of β-actin, TNF-α, and IL-10 normalized by β-actin (n = 6/group). **P < 0.01, ***P < 0.001 compared with wild-type mice that received sham surgery, ##P < 0.01 compared with wild-type mice that received SNI surgery. $$P < 0.01, $$$P < 0.001 compared with wild-type mice that received SNI surgery. Scale bar (A) = 50 µm. DMSO = dimethyl sulfoxide.

Fig. 5.

Activation of astrocyte and microglia in the spinal cord was enhanced in liver X receptor (LXR) α knockout ([–/–]) mice after spared nerve injury (SNI), and GW3965 inhibited activation of astrocytes and microglia, down-regulated interleukin (IL) 1β and tumor necrosis factor (TNF) α, and up-regulated IL-10 expression in the spinal dorsal horn in wild-type but not in LXRα (–/–) mice. (A) Expression of glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1) in six different groups is shown. (B, C) The histogram shows the summary data of the GFAP- or Iba1-positive area in different groups (n = 6/group). (D through F) Protein levels of IL-1β, TNF-α, IL-10, and β-actin from samples of ipsilateral spinal dorsal horn in eight groups. The histogram shows the quantification of β-actin, TNF-α, and IL-10 normalized by β-actin (n = 6/group). **P < 0.01, ***P < 0.001 compared with wild-type mice that received sham surgery, ##P < 0.01 compared with wild-type mice that received SNI surgery. $$P < 0.01, $$$P < 0.001 compared with wild-type mice that received SNI surgery. Scale bar (A) = 50 µm. DMSO = dimethyl sulfoxide.

Close modal

Intrathecal injection of T0901317 twenty-one days after spared nerve injury did not affect the activation of microglia and astrocytes (Supplemental Digital Content 2, http://links.lww.com/ALN/B470). Compared with the vehicle-treated group (Supplemental Digital Content 2A, http://links.lww.com/ALN/B470), the positive area of Iba1 (Supplemental Digital Content 2B, http://links.lww.com/ALN/B470) and GFAP (Supplemental Digital Content 2E, http://links.lww.com/ALN/B470) 1 h after T0901317 treatment was not changed. The same results were observed 2 h after T0901317 treatment (Supplemental Digital Content 2C and 2F, http://links.lww.com/ALN/B470).

T0901317 Up-regulated Neurosteroid-related Enzymes in Spared Nerve Injury Rats, and the Inhibitory Effect of T0901317 Was Blocked by Aminoglutethimide

It has been shown that LXRs function via the promotion of neurosteroidogenesis.23  Here first we tested whether endogenous neurosteroid played a role in pain hypersensitivity and neuroinflammation after spared nerve injury. As shown in Supplemental Digital Content 3 (http://links.lww.com/ALN/B471), intraperitoneal injection of neurosteroid synthesis inhibitor aminoglutethimide (10 min after spared nerve injury and once every third day) could facilitate the development of mechanical allodynia but did not affect the maintenance of mechanical allodynia. In aminoglutethimide-treated spared nerve injury rats, mechanical allodynia in the ipsilateral hind paw developed 1 day after spared nerve injury, whereas in the vehicle-treatment group, mechanical allodynia did not develop until 2 days after spared nerve injury.

We next tested whether neurosteroidogenesis was involved in the inhibitory effect of LXR agonists on mechanical allodynia. As shown in figure 6A, expression of cytochrome P450 side chain cleavage ([P450scc] the enzyme responsible of the conversion of cholesterol into pregnenolone) was significantly increased 7 days after spared nerve injury and remained at a significantly high level at day 14, whereas steroidogenic acute regulatory protein ([StAR] molecules involved in cholesterol shuttling into the mitochondria) was increased at 4 days after spared nerve injury. The expression of P450scc was not changed, whereas the expression of StAR was up-regulated (fig. 6B) after T0901317 treatment. The expression of StAR and P450scc could only be up-regulated 7 days after spared nerve injury in the wild-type mice but not in LXRα (–/–) mice (fig. 6C). Pretreatment with aminoglutethimide (30 min before T0901317) could completely block the effect of T0901317 on the decrease of the 50% paw withdrawal threshold induced by spared nerve injury (fig. 6D). After T0901317 treatment, the expression of StAR was located mainly in neurons (fig. 6E) and microglia (fig. 6F) in the spinal gray matter, but not in astrocytes (fig. 6G), whereas in the white matter, StAR was located mainly in oligodendrocytes (fig. 6H) and microglia (fig. 6I) but not in astrocytes (fig. 6J). A similar expression pattern of StAR was observed from rats that received spared nerve injury surgery 9 days before (data not shown).

Fig. 6.

T0901317 up-regulated neurosteroid-related enzymes in spared nerve injury (SNI) rats, and the inhibitory effect of T0901317 was blocked by aminoglutethimide. (A) cytochrome P450 side chain cleavage (P450scc) and steroidogenic acute regulatory protein (StAR) in the ipsilateral dorsal lumbar spinal cord were up-regulated after SNI (n = 5/group). *P < 0.05, **P < 0.01, ***P < 0.001 compared with sham group. (B) T0901317 down-regulated the expression of P450scc and StAR. **P < 0.01, ***P < 0.001 compared with sham group; ##P < 0.01 compared with vehicle-treated SNI group. (C) The bands show the expression of P450scc, StAR, and β-actin in four different groups (n = 4/group). **P < 0.01, ***P < 0.001 compared with wild-type mice that received sham surgery; $$P < 0.01 compared with liver X receptor (LXR) α knockout mice that received SNI surgery. (D) Aminoglutethimide abolished the effects of T0901317 (n = 5 to 6/group). Arrow indicates the time of T0901317 injection. **P < 0.01 compared with before T0901317. ##P < 0.01 compared with dimethyl sulfoxide (DMSO)– and T0901317-treated group. (E through J) The cell types that expressed StAR in the gray and white matter. Scale bar (E through J) = 50 µm. GFAP = glial fibrillary acidic protein; Iba1 = ionized calcium binding adaptor molecule 1; NeuN = neuron specific nuclear protein; Nogo-A = neurite outgrowth inhibitor A.

Fig. 6.

T0901317 up-regulated neurosteroid-related enzymes in spared nerve injury (SNI) rats, and the inhibitory effect of T0901317 was blocked by aminoglutethimide. (A) cytochrome P450 side chain cleavage (P450scc) and steroidogenic acute regulatory protein (StAR) in the ipsilateral dorsal lumbar spinal cord were up-regulated after SNI (n = 5/group). *P < 0.05, **P < 0.01, ***P < 0.001 compared with sham group. (B) T0901317 down-regulated the expression of P450scc and StAR. **P < 0.01, ***P < 0.001 compared with sham group; ##P < 0.01 compared with vehicle-treated SNI group. (C) The bands show the expression of P450scc, StAR, and β-actin in four different groups (n = 4/group). **P < 0.01, ***P < 0.001 compared with wild-type mice that received sham surgery; $$P < 0.01 compared with liver X receptor (LXR) α knockout mice that received SNI surgery. (D) Aminoglutethimide abolished the effects of T0901317 (n = 5 to 6/group). Arrow indicates the time of T0901317 injection. **P < 0.01 compared with before T0901317. ##P < 0.01 compared with dimethyl sulfoxide (DMSO)– and T0901317-treated group. (E through J) The cell types that expressed StAR in the gray and white matter. Scale bar (E through J) = 50 µm. GFAP = glial fibrillary acidic protein; Iba1 = ionized calcium binding adaptor molecule 1; NeuN = neuron specific nuclear protein; Nogo-A = neurite outgrowth inhibitor A.

Close modal

Activation of LXRs Reversed Spared Nerve Injury–induced Up-regulation of LXRα, and Pretreatment with Aminoglutethimide Blocked the Effect

T0901317 decreased the upregulation of LXRα induced by spared nerve injury (fig. 7). Again, the effect was prevented by aminoglutethimide (fig. 7). T0901317 alone had no effect on the expression of LXRα in sham rats.

Fig. 7.

T0901317 down-regulated liver X receptor (LXR) α expression in the spinal dorsal horn, and this effect was prevented by aminoglutethimide. Bands show the expression of LXRα and β-actin in the five groups, as indicated. Histograms show the expression of LXRα and β-actin in the above groups (n = 5/group). ***P < 0.001 compared with dimethyl sulfoxide (DMSO)–treated sham group. ###P <0.001 compared with DMSO-treated spared nerve injury (SNI) group. $$$P < 0.001 compared with T0901317-treated SNI group.

Fig. 7.

T0901317 down-regulated liver X receptor (LXR) α expression in the spinal dorsal horn, and this effect was prevented by aminoglutethimide. Bands show the expression of LXRα and β-actin in the five groups, as indicated. Histograms show the expression of LXRα and β-actin in the above groups (n = 5/group). ***P < 0.001 compared with dimethyl sulfoxide (DMSO)–treated sham group. ###P <0.001 compared with DMSO-treated spared nerve injury (SNI) group. $$$P < 0.001 compared with T0901317-treated SNI group.

Close modal

T0901317 Inhibited Activation of Astrocytes and Microglia, Decreased IL-1β, and Increased IL-10 Production in the Spinal Dorsal Horn, and the Effects Were Prevented by Aminoglutethimide

Expression of both GFAP and Iba1 in the ipsilateral lumbar spinal dorsal horn (fig. 8A) was significantly higher at day 9 in dimethyl sulfoxide–treated spared nerve injury rats than that in dimethyl sulfoxide–treated sham rats. In addition, the expression of GFAP and Iba1 in aminoglutethimide- and T0901317-treated spared nerve injury rats was higher than that of T0901317-treated spared nerve injury rats, and there was no difference from the dimethyl sulfoxide–treated spared nerve injury group. T0901317 alone had no effect on the expression of GFAP and Iba1 in sham rats.

Fig. 8.

Aminoglutethimide prevented the effects of T0901317 on glial cells activation, interleukin (IL) 1β, and IL-10 expression in the spinal dorsal horn. (A) Representative bands showing the expression of glial fibrillary acidic protein (GFAP), (ionized calcium binding adaptor molecule 1) Iba1, and β-actin in 5 different groups are shown above each histogram (n = 5/group). ***P < 0.001, ###P < 0.001, $$$P < 0.001, compared with dimethyl sulfoxide (DMSO)–treated sham group, DMSO-treated spared nerve injury (SNI) group, and T0901317-treated SNI group, respectively. (B) The bands and histogram show the expression of IL-1β, IL-10, and β-actin in the above five groups and the quantification of IL-1β and IL-10 normalized by β-actin (n = 6/group). *P < 0.05, ***P < 0.001 compared with DMSO-treated sham group; #P < 0.05, ##P < 0.01 compared with DMSO-treated SNI group. $P < 0.05, $$$P < 0.001 compared with T0901317-treated SNI group. (C through N) Cell types that expressed IL-1β and IL-10 in the gray and white matter. Scale bar (B through M) = 50 µm. NeuN = neuron specific nuclear protein; Nogo-A = Neurite outgrowth inhibitor A.

Fig. 8.

Aminoglutethimide prevented the effects of T0901317 on glial cells activation, interleukin (IL) 1β, and IL-10 expression in the spinal dorsal horn. (A) Representative bands showing the expression of glial fibrillary acidic protein (GFAP), (ionized calcium binding adaptor molecule 1) Iba1, and β-actin in 5 different groups are shown above each histogram (n = 5/group). ***P < 0.001, ###P < 0.001, $$$P < 0.001, compared with dimethyl sulfoxide (DMSO)–treated sham group, DMSO-treated spared nerve injury (SNI) group, and T0901317-treated SNI group, respectively. (B) The bands and histogram show the expression of IL-1β, IL-10, and β-actin in the above five groups and the quantification of IL-1β and IL-10 normalized by β-actin (n = 6/group). *P < 0.05, ***P < 0.001 compared with DMSO-treated sham group; #P < 0.05, ##P < 0.01 compared with DMSO-treated SNI group. $P < 0.05, $$$P < 0.001 compared with T0901317-treated SNI group. (C through N) Cell types that expressed IL-1β and IL-10 in the gray and white matter. Scale bar (B through M) = 50 µm. NeuN = neuron specific nuclear protein; Nogo-A = Neurite outgrowth inhibitor A.

Close modal

In dimethyl sulfoxide–treated spared nerve injury rats (9 days after spared nerve injury), the expression of IL-1β was increased, whereas IL-10 was decreased in the ipsilateral spinal dorsal horn. The increased IL-1β and decreased IL-10 produced by spared nerve injury were suppressed by T0901317. The effect of T0901317 on IL-1β (fig. 8B) and IL-10 (fig. 8B) was completely blocked by aminoglutethimide. After T0901317 treatment, the expression of IL-1β was located mainly in neurons and astrocytes (fig. 8, C and D) but not in microglia (fig. 8E) in spinal gray matter. In the white matter, IL-1β was located mainly in oligodendrocytes (fig. 8F), with a lesser extent in astrocytes (fig. 8G) and microglia (fig. 8H). Only neurons (fig. 8I), but not astrocytes (fig. 8J) or microglia (fig. 8K), express IL-10 in the gray matter, whereas in the white matter, IL-10 was expressed mainly in oligodendrocytes (fig. 8L) but not in astrocytes (fig. 8M) or in microglia (fig. 8N). The expression pattern of IL-1β and IL-10 was consistent with that obtained from rats that received spared nerve injury surgery 9 days before (data not shown).

In the present study, we showed that LXRα was up-regulated in neurons and oligodendrocytes in the spinal dorsal horn after spared nerve injury, and intrathecal injection of LXR agonists GW3965 or T0901317 could alleviate mechanical allodynia. GW3965 could inhibit the activation of microglia and astrocytes and rebalance the expression of proinflammatory and antiinflammatory cytokines. All of these above effects of LXR agonists were abolished by LXRα mutation. Furthermore, more glial cells were activated and more IL-1β was produced in the spinal dorsal horn after spared nerve injury in LXRα (–/–) mice. Aminoglutethimide, a neurosteroid synthesis inhibitor, could completely block the effects of T0901317 on the expression of LXRα, the activation of microglia and astrocytes, the up-regulation of IL-1β, and the down-regulation of IL-10 induced by spared nerve injury. Moreover, after T0901317 treatment, the expressions of StAR, IL-1β, and IL-10 were all located mainly in neurons and oligodendrocytes, indicating that LXR agonists can exert a stimulating effect by activating LXRα located in both neurons and oligodendrocytes and thus promoting the synthesis of neurosteroids to counteract neuropathic pain.

Up-regulation of LXRα Is Protective against Neuronal Damage

Our previous work had shown that translocator protein (18kDa) was up-regulated in spinal dorsal horn after lumbar 5 spinal nerve ligation and that a translocator protein agonist could reverse neuropathic pain behaviors, including mechanical allodynia and thermal hyperalgesia.24  Similarly, here we showed that LXRα was up-regulated in the bilateral spinal dorsal horn after spared nerve injury and that a single dose of LXR agonists (either T0901317 or GW3965) inhibited mechanical allodynia. The inhibitory effect of LXR agonists was abolished by LXRα mutation. Moreover, T0901317 could decrease the up-regulated LXRα while inhibiting mechanical allodynia, and the effect of T0901317 on LXRα was again abrogated by aminoglutethimide. These results suggest that the function of LXRα is in trying to inhibit neuropathic pain. Once pain was alleviated by LXRα agonists, the expression of LXRα would decrease. After aminoglutethimide treatment, because the animals kept experiencing pain, LXRα overexpression was kept there to counteract the pain state. This also suggests that, similar to translocator protein, LXRα acts as a self-protecting mechanism in animals experiencing pain. Previous studies have demonstrated the potent neuroprotective effect of LXRs in many disease models. For example, it has been reported that LXRs protect against a loss of dopaminergic neurons and of dopaminergic fibers projecting to the striatum,25  that LXRα reduces cardiac hypertrophy by improving glucose uptake and use,26  and that activation of LXRs protects N-methyl-d-aspartate–induced inner retinal damage.27  It was also reported that LXR and its target gene adenosine triphosphate–binding cassette transporters could modulate diabetic retinopathy outcome in streptozotocin-induced diabetes mellitus28  and in experimental autoimmune uveitis.29  As a key sensor of cholesterol, LXRs react to the increased cellular level of cholesterol, leading to transcription of a series of genes and thus protection for the cells. Therefore, it is possible that cholesterol overload is the common reason for all the above diseases, and LXRs are the common target to ameliorate these diseases. The long-lasting effect of a single injection of an LXR agonist suggests that promoting the function of LXRα is an effective way to counteract neuropathic pain. Additional experiments should be performed to test whether LXRβ plays a role in neuropathic pain.

In the present study, we found that in the lumbar spinal cords of sham-operated animals from the wild-type group, the expression of IL-10, IL-1β, and TNF-α was at the same level as from LXRα (–/–) mice, and these animals are not in pain (data not shown). These results, together with the result showing that T0901317 had no effect on the paw withdrawal incidence in the sham-operated rats, suggest that LXRαs do not exert effects under normal conditions. Moreover, the expression of IL-1β was increased after spared nerve injury in LXRα (–/–) mice compared with the wild-type mice, whereas the expression of IL-10 and TNF-α was not changed. These results suggest that, under neuropathic pain conditions, LXRα mutation only affects expression of part of the cytokines.

Mechanisms Show That LXR Agonists Inhibit Mechanical Allodynia

Activation of the LXRs may benefit several neurologic disorders by modulating functions of neurons directly or indirectly. LXR activation improves axonal regeneration in patients with stroke,30  protects dopaminergic neurons in Parkinson disease,25  and reduces the loss of cholinergic neurons in Alzheimer disease.31  The LXR ligand could reduce markers of neuroinflammation, such as nitric oxide synthase, and reverse microglia from activated states to resting states.32  In accordance with these studies, we found that T0901317 inhibited the activation of glial cells in the spinal dorsal horn. In addition, the expression of TNF-α and IL-1β was down-regulated and IL-10 was up-regulated by T0901317. By rebalancing the expression of proinflammatory and antiinflammatory cytokines, T0901317 may optimize the environment in which the neurons live. More importantly, the inhibitory effect of the LXR agonist could be abolished in LXRα (–/–) mice, suggesting that LXRα plays an important role in mediating the inhibitory effect of GW3965 and T0901317. Two days after LXR treatment, LXRα, StAR, IL-1β, and IL-10 were all located in neurons and oligodendrocytes, indicating that the LXR agonist can exert a stimulating effect by activation of LXRα located in both neurons and oligodendrocytes. Because the effects of T0901317 could be blocked by neuroactive steroid synthesis inhibitor aminoglutethimide, LXR agonists may first increase the synthesis of neurosteroids and then decrease the synthesis of IL-1β, hence inhibiting the glial cell activation and at last the excitability of neurons. IL-10, an antiinflammatory cytokine, could inhibit the production of many proinflammatory cytokines, including IL-1β, TNF-α, and interleukin 6.33,34  It is probable that LXR agonists first increase the synthesis of IL-10 and then decrease the production of proinflammatory cytokines. These results also indicate that, after nerve injury, neurons and oligodendrocytes can combine hands to counteract the abnormal state. In the present study, LXR agonists were injected via the intrathecal routine, which may affect both lumbar dorsal root ganglias and spinal cord. Because LXRα is also expressed in dorsal root ganglia neurons, LXRα here may also be involved in mediating the inhibitory effect of LXR agonists.

Previous data indicate that neuroactive steroids, including pregnenolone, progesterone, and allopregnanolone, are synthesized and actively metabolized in the central and peripheral nervous system and exert a neuroprotective effect in states of neuropathic pain induced by lumbar 5 spinal nerve ligation,35  chronic constriction injury,36  or spinal cord injury.37  We have shown previously that a single application of Ro5-4864, a specific translocator protein agonist,24  and pregnenolone, a neurosteroid precursor,38  inhibits the established neuropathic pain behaviors induced by lumbar 5 spinal nerve ligation. In the current study, we found that endogenous neurosteroids could postpone the development of mechanical allodynia. A line of study has reported that a LXR agonist increases levels of neuroactive steroid and prevents diabetes-induced peripheral neuropathy.23  Recently, it has been reported that LXRs directly modulate StAR expression in the adrenal gland,39  a transfer protein regulating cholesterol shuttling into mitochondria, which is a key step in the initiation of steroid hormone synthesis. Consistently, in the present study, we found that T0901317 up-regulated expression of StAR. These results suggest that activation of LXRs stimulates the synthesis of neuroactive steroids. In addition, we found that the effects of T0901317 on mechanical allodynia, activation of glial cells, and expression of cytokines could be prevented by a neurosteroid synthesis inhibitor. Therefore, T0901317 may function via neurosteroids. Improving the local neurosteroid synthesis by activation of LXRs may be a valid way to treat neuropathic pain.

In conclusion, the present study demonstrated that a novel role of spinal LXRα in neuropathic pain, that is, activation of LXRα at the early time after nerve injury, can inhibit mechanical allodynia via modulating the neuroinflammatory responses.

Supported by grants from the National Natural Science Foundation (Beijing, People’s Republic of China, Nos. U1201223, 81200856, and 81471250); Fundamental Research Funds for the Central Universities (Beijing, People’s Republic of China, No. 13ykpy05); Nature Science Foundation of Guangdong Province of China (Guangzhou, People’s Republic of China, No. 2014A030313029); Scientific Research Foundation of Guangdong Province of China (Guangzhou, People’s Republic of China, No. 2016A020215035); and by scholarships from the Chinese Scholarship Council (Beijing, People’s Republic of China). All grants were awarded to Dr. Wei except No. U1201223 which was awarded to Dr. Liu.

The authors declare no competing interests.

1.
Baron
R
:
Mechanisms of disease: Neuropathic pain–A clinical perspective.
Nat Clin Pract Neurol
2006
;
2
:
95
106
2.
Viennois
E
,
Mouzat
K
,
Dufour
J
,
Morel
L
,
Lobaccaro
JM
,
Baron
S
:
Selective liver X receptor modulators (SLiMs): What use in human health?
Mol Cell Endocrinol
2012
;
351
:
129
41
3.
Zelcer
N
,
Tontonoz
P
:
Liver X receptors as integrators of metabolic and inflammatory signaling.
J Clin Invest
2006
;
116
:
607
14
4.
Repa
JJ
,
Mangelsdorf
DJ
:
The role of orphan nuclear receptors in the regulation of cholesterol homeostasis.
Annu Rev Cell Dev Biol
2000
;
16
:
459
81
5.
Guillem-Llobat
P
,
Íñiguez
MA
:
Inhibition of lipopolysaccharide-induced gene expression by liver X receptor ligands in macrophages involves interference with early growth response factor 1.
Prostaglandins Leukot Essent Fatty Acids
2015
;
96
:
37
49
6.
Paterniti
I
,
Genovese
T
,
Mazzon
E
,
Crisafulli
C
,
Di Paola
R
,
Galuppo
M
,
Bramanti
P
,
Cuzzocrea
S
:
Liver X receptor agonist treatment regulates inflammatory response after spinal cord trauma.
J Neurochem
2010
;
112
:
611
24
7.
Clark
AK
,
Gentry
C
,
Bradbury
EJ
,
McMahon
SB
,
Malcangio
M
:
Role of spinal microglia in rat models of peripheral nerve injury and inflammation.
Eur J Pain
2007
;
11
:
223
30
8.
Ji
XT
,
Qian
NS
,
Zhang
T
,
Li
JM
,
Li
XK
,
Wang
P
,
Zhao
DS
,
Huang
G
,
Zhang
L
,
Fei
Z
,
Jia
D
,
Niu
L
:
Spinal astrocytic activation contributes to mechanical allodynia in a rat chemotherapy-induced neuropathic pain model.
PLoS One
2013
;
8
:
e60733
9.
Clark
AK
,
D’Aquisto
F
,
Gentry
C
,
Marchand
F
,
McMahon
SB
,
Malcangio
M
:
Rapid co-release of interleukin 1beta and caspase 1 in spinal cord inflammation.
J Neurochem
2006
;
99
:
868
80
10.
Zhou
LJ
,
Yang
T
,
Wei
X
,
Liu
Y
,
Xin
WJ
,
Chen
Y
,
Pang
RP
,
Zang
Y
,
Li
YY
,
Liu
XG
:
Brain-derived neurotrophic factor contributes to spinal long-term potentiation and mechanical hypersensitivity by activation of spinal microglia in rat.
Brain Behav Immun
2011
;
25
:
322
34
11.
Wei
XH
,
Na
XD
,
Liao
GJ
,
Chen
QY
,
Cui
Y
,
Chen
FY
,
Li
YY
,
Zang
Y
,
Liu
XG
:
The up-regulation of IL-6 in DRG and spinal dorsal horn contributes to neuropathic pain following L5 ventral root transection.
Exp Neurol
2013
;
241
:
159
68
12.
Mika
J
:
Modulation of microglia can attenuate neuropathic pain symptoms and enhance morphine effectiveness.
Pharmacol Rep
2008
;
60
:
297
307
13.
Milligan
ED
,
Sloane
EM
,
Langer
SJ
,
Hughes
TS
,
Jekich
BM
,
Frank
MG
,
Mahoney
JH
,
Levkoff
LH
,
Maier
SF
,
Cruz
PE
,
Flotte
TR
,
Johnson
KW
,
Mahoney
MM
,
Chavez
RA
,
Leinwand
LA
,
Watkins
LR
:
Repeated intrathecal injections of plasmid DNA encoding interleukin-10 produce prolonged reversal of neuropathic pain.
Pain
2006
;
126
:
294
308
14.
Shen
KF
,
Zhu
HQ
,
Wei
XH
,
Wang
J
,
Li
YY
,
Pang
RP
,
Liu
XG
:
Interleukin-10 down-regulates voltage gated sodium channels in rat dorsal root ganglion neurons.
Exp Neurol
2013
;
247
:
466
75
15.
Zimmermann
M
:
Ethical guidelines for investigations of experimental pain in conscious animals.
Pain
1983
;
16
:
109
10
16.
Schultz
JR
,
Tu
H
,
Luk
A
,
Repa
JJ
,
Medina
JC
,
Li
L
,
Schwendner
S
,
Wang
S
,
Thoolen
M
,
Mangelsdorf
DJ
,
Lustig
KD
,
Shan
B
:
Role of LXRs in control of lipogenesis.
Genes Dev
2000
;
14
:
2831
8
17.
Collins
JL
,
Fivush
AM
,
Watson
MA
,
Galardi
CM
,
Lewis
MC
,
Moore
LB
,
Parks
DJ
,
Wilson
JG
,
Tippin
TK
,
Binz
JG
,
Plunket
KD
,
Morgan
DG
,
Beaudet
EJ
,
Whitney
KD
,
Kliewer
SA
,
Willson
TM
:
Identification of a nonsteroidal liver X receptor agonist through parallel array synthesis of tertiary amines.
J Med Chem
2002
;
45
:
1963
6
18.
Decosterd
I
,
Woolf
CJ
:
Spared nerve injury: An animal model of persistent peripheral neuropathic pain.
Pain
2000
;
87
:
149
58
19.
Chaplan
SR
,
Bach
FW
,
Pogrel
JW
,
Chung
JM
,
Yaksh
TL
:
Quantitative assessment of tactile allodynia in the rat paw.
J Neurosci Methods
1994
;
53
:
55
63
20.
Eschenfelder
S
,
Häbler
HJ
,
Jänig
W
:
Dorsal root section elicits signs of neuropathic pain rather than reversing them in rats with L5 spinal nerve injury.
Pain
2000
;
87
:
213
9
21.
Wei
XH
,
Zang
Y
,
Wu
CY
,
Xu
JT
,
Xin
WJ
,
Liu
XG
:
Peri-sciatic administration of recombinant rat TNF-alpha induces mechanical allodynia via upregulation of TNF-alpha in dorsal root ganglia and in spinal dorsal horn: The role of NF-kappa B pathway.
Exp Neurol
2007
;
205
:
471
84
22.
Kojetin
DJ
,
Burris
TP
:
REV-ERB and ROR nuclear receptors as drug targets.
Nat Rev Drug Discov
2014
;
13
:
197
216
23.
Cermenati
G
,
Giatti
S
,
Cavaletti
G
,
Bianchi
R
,
Maschi
O
,
Pesaresi
M
,
Abbiati
F
,
Volonterio
A
,
Saez
E
,
Caruso
D
,
Melcangi
RC
,
Mitro
N
:
Activation of the liver X receptor increases neuroactive steroid levels and protects from diabetes-induced peripheral neuropathy.
J Neurosci
2010
;
30
:
11896
901
24.
Wei
XH
,
Wei
X
,
Chen
FY
,
Zang
Y
,
Xin
WJ
,
Pang
RP
,
Chen
Y
,
Wang
J
,
Li
YY
,
Shen
KF
,
Zhou
LJ
,
Liu
XG
:
The upregulation of translocator protein (18 kDa) promotes recovery from neuropathic pain in rats.
J Neurosci
2013
;
33
:
1540
51
25.
Dai
YB
,
Tan
XJ
,
Wu
WF
,
Warner
M
,
Gustafsson
:
Liver X receptor β protects dopaminergic neurons in a mouse model of Parkinson disease.
Proc Natl Acad Sci U|S|A
2012
;
109
:
13112
7
26.
Cannon
MV
,
Silljé
HH
,
Sijbesma
JW
,
Vreeswijk-Baudoin
I
,
Ciapaite
J
,
van der Sluis
B
,
van Deursen
J
,
Silva
GJ
,
de Windt
LJ
,
Gustafsson
,
van der Harst
P
,
van Gilst
WH
,
de Boer
RA
:
Cardiac LXRα protects against pathological cardiac hypertrophy and dysfunction by enhancing glucose uptake and utilization.
EMBO Mol Med
2015
;
7
:
1229
43
27.
Zheng
S
,
Yang
H
,
Chen
Z
,
Zheng
C
,
Lei
C
,
Lei
B
:
Activation of liver X receptor protects inner retinal damage induced by N-methyl-D-aspartate.
Invest Ophthalmol Vis Sci
2015
;
56
:
1168
80
28.
Hazra
S
,
Rasheed
A
,
Bhatwadekar
A
,
Wang
X
,
Shaw
LC
,
Patel
M
,
Caballero
S
,
Magomedova
L
,
Solis
N
,
Yan
Y
,
Wang
W
,
Thinschmidt
JS
,
Verma
A
,
Li
Q
,
Levi
M
,
Cummins
CL
,
Grant
MB
:
Liver X receptor modulates diabetic retinopathy outcome in a mouse model of streptozotocin-induced diabetes.
Diabetes
2012
;
61
:
3270
9
29.
Yang
H
,
Zheng
S
,
Qiu
Y
,
Yang
Y
,
Wang
C
,
Yang
P
,
Li
Q
,
Lei
B
:
Activation of liver X receptor alleviates ocular inflammation in experimental autoimmune uveitis.
Invest Ophthalmol Vis Sci
2014
;
55
:
2795
804
30.
Chen
J
,
Zacharek
A
,
Cui
X
,
Shehadah
A
,
Jiang
H
,
Roberts
C
,
Lu
M
,
Chopp
M
:
Treatment of stroke with a synthetic liver X receptor agonist, TO901317, promotes synaptic plasticity and axonal regeneration in mice.
J Cereb Blood Flow Metab
2010
;
30
:
102
9
31.
Cui
W
,
Sun
Y
,
Wang
Z
,
Xu
C
,
Peng
Y
,
Li
R
:
Liver X receptor activation attenuates inflammatory response and protects cholinergic neurons in APP/PS1 transgenic mice.
Neuroscience
2012
;
210
:
200
10
32.
Repa
JJ
,
Li
H
,
Frank-Cannon
TC
,
Valasek
MA
,
Turley
SD
,
Tansey
MG
,
Dietschy
JM
:
Liver X receptor activation enhances cholesterol loss from the brain, decreases neuroinflammation, and increases survival of the NPC1 mouse.
J Neurosci
2007
;
27
:
14470
80
33.
Ouyang
W
,
Rutz
S
,
Crellin
NK
,
Valdez
PA
,
Hymowitz
SG
:
Regulation and functions of the IL-10 family of cytokines in inflammation and disease.
Annu Rev Immunol
2011
;
29
:
71
109
34.
He
Z
,
Guo
Q
,
Xiao
M
,
He
C
,
Zou
W
:
Intrathecal lentivirus-mediated transfer of interleukin-10 attenuates chronic constriction injury-induced neuropathic pain through modulation of spinal high-mobility group box 1 in rats.
Pain Physician
2013
;
16
:
E615
25
35.
Liu
X
,
Li
W
,
Dai
L
,
Zhang
T
,
Xia
W
,
Liu
H
,
Ma
K
,
Xu
J
,
Jin
Y
:
Early repeated administration of progesterone improves the recovery of neuropathic pain and modulates spinal 18kDa-translocator protein (TSPO) expression.
J Steroid Biochem Mol Biol
2014
;
143
:
130
40
36.
Jarahi
M
,
Sheibani
V
,
Safakhah
HA
,
Torkmandi
H
,
Rashidy-Pour
A
:
Effects of progesterone on neuropathic pain responses in an experimental animal model for peripheral neuropathy in the rat: A behavioral and electrophysiological study.
Neuroscience
2014
;
256
:
403
11
37.
Coronel
MF
,
Labombarda
F
,
De Nicola
AF
,
González
SL
:
Progesterone reduces the expression of spinal cyclooxygenase-2 and inducible nitric oxide synthase and prevents allodynia in a rat model of central neuropathic pain.
Eur J Pain
2014
;
18
:
348
59
38.
Chen
SL
,
Zang
Y
,
Zheng
WH
,
Wei
XH
,
Liu
XG
:
Inhibition of neuropathic pain by a single intraperitoneal injection of diazepam in the rat: Possible role of neurosteroids.
Chin J Physiol
2016
;
59
:
9
20
39.
Cummins
CL
,
Volle
DH
,
Zhang
Y
,
McDonald
JG
,
Sion
B
,
Lefrançois-Martinez
AM
,
Caira
F
,
Veyssière
G
,
Mangelsdorf
DJ
,
Lobaccaro
JM
:
Liver X receptors regulate adrenal cholesterol balance.
J Clin Invest
2006
;
116
:
1902
12