Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2013

Open Access 01.12.2013 | Research

Bilateral elevation of interleukin-6 protein and mRNA in both lumbar and cervical dorsal root ganglia following unilateral chronic compression injury of the sciatic nerve

verfasst von: Petr Dubový, Václav Brázda, Ilona Klusáková, Ivana Hradilová-Svíženská

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2013

Abstract

Background

Current research implicates interleukin (IL)-6 as a key component of the nervous-system response to injury with various effects.

Methods

We used unilateral chronic constriction injury (CCI) of rat sciatic nerve as a model for neuropathic pain. Immunofluorescence, ELISA, western blotting and in situ hybridization were used to investigate bilateral changes in IL-6 protein and mRNA in both lumbar (L4-L5) and cervical (C7-C8) dorsal root ganglia (DRG) following CCI. The operated (CCI) and sham-operated (sham) rats were assessed after 1, 3, 7, and 14 days. Withdrawal thresholds for mechanical hyperalgesia and latencies for thermal hyperalgesia were measured in both ipsilateral and contralateral hind and fore paws.

Results

The ipsilateral hind paws of all CCI rats displayed a decreased threshold of mechanical hyperalgesia and withdrawal latency of thermal hyperalgesia, while the contralateral hind and fore paws of both sides exhibited no significant changes in mechanical or thermal sensitivity. No significant behavioral changes were found in the hind and fore paws on either side of the sham rats, except for thermal hypersensitivity, which was present bilaterally at 3 days. Unilateral CCI of the sciatic nerve induced a bilateral increase in IL-6 immunostaining in the neuronal bodies and satellite glial cells (SGC) surrounding neurons of both lumbar and cervical DRG, compared with those of naive control rats. This bilateral increase in IL-6 protein levels was confirmed by ELISA and western blotting. More intense staining for IL-6 mRNA was detected in lumbar and cervical DRG from both sides of rats following CCI. The DRG removed from sham rats displayed a similar pattern of staining for IL-6 protein and mRNA as found in naive DRG, but there was a higher staining intensity in SGC.

Conclusions

Bilateral elevation of IL-6 protein and mRNA is not limited to DRG homonymous to the injured nerve, but also extended to DRG that are heteronymous to the injured nerve. The results for IL-6 suggest that the neuroinflammatory reaction of DRG to nerve injury is propagated alongside the neuroaxis from the lumbar to the remote cervical segments. This is probably related to conditioning of cervical DRG neurons to injury.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

PD conceived, designed and coordinated the study and wrote the manuscript. VB conceived, designed, and coordinated the western blot and in situ hybridization analyses. IK and IS conceived, designed and carried out the experiments, and participated in acquiring and analyzing the presented data. All authors gave final approval to the version to be published.

Background

Neuropathic pain has recently been defined as ‘pain arising as a direct consequence of a lesion or disease affecting the somatosensory system’ [1] and is therefore not directly associated with nociceptive input. Peripheral neuropathic pain, manifested as spontaneous pain and hyperalgesia, arises as a result of various forms of peripheral nerve damage, such as traumatic nerve injury, or neuropathy associated with diabetes or HIV infection [2, 3].
There is compelling evidence indicating that hyperalgesia and ongoing pain due to peripheral nerve injury are associated with excitability of [4] and cellular and molecular changes in dorsal root ganglia (DRG), including proliferation and activation of satellite glial cells (SGC) [5], invasion of macrophages [6], and upregulation and downregulation of genes and proteins [7, 8].
Pro-inflammatory and anti-inflammatory cytokines contribute to both induction and maintenance of neuropathic pain derived from cellular and molecular changes in the DRG [9, 10]. Interleukin (IL)-6 is a member of the family of cytokines collectively termed ‘the interleukin-6-type cytokines’, which have diverse functions throughout the body. A growing body of evidence implicates IL-6 as a key component in the response of the nervous system to injury. For example, IL-6 is involved in promoting neuronal survival and protection against neuronal damage [11, 12] and also in modulating pain [13, 14].
In response to sciatic nerve transection, IL-6 protein and mRNA levels were found to be raised in medium to large sensory neurons 2 to 4 days after such damage in the ipsilateral, but not the contralateral, DRG homonymous to the injured nerve. By contrast, a nerve-constriction model induced lower concentrations of IL-6 protein and mRNA in DRG neurons, but both persisted longer than in a nerve-transection model. Presence of IL-6 in the nerve-constriction model of neuropathic pain correlated well with the duration of hypersensitivity [15, 16].
A growing body of evidence indicates that unilateral nerve injury results in bilateral cellular and molecular changes in the nerve structures [17, 18], and in bilateral changes indicated by behavioral tests [19]. In addition, neighboring uninjured DRG display changes after lesion of non-associated nerves [20, 21].
The aim of the present study was to investigate quantitative alterations in IL-6 protein and mRNA levels following unilateral chronic constriction injury (CCI) of the sciatic nerve in both ipsilateral and contralateral DRG at L4-L5 and C7-C8 levels.

Methods

Animals and surgical procedures

Procedures were performed in accordance with protocols approved by the Animal Investigation Committee of the Faculty of Medicine, Brno, Czech Republic, and followed ethical guidance [22].
All experimental procedures were carried out under sterile conditions by the same person. The experiments were performed using 159 adult male Wistar rats 250–300 g in weight (Anlab, Brno, Czech Republic). The animals were housed on a 12 hour light/12 hour dark cycle at a temperature of 22 to 24°C, under specific pathogen-free conditions in the animal housing facility of Masaryk University. Sterilized standard rodent food and water were available ad libitum.
Surgical procedures were performed under deep anesthesia with a mixture of equal volumes of intraperitoneal (IP) ketamine 40 mg/ml and xylazine 4 mg/ml (Bioveta a.s., Czech Republic) (0.2 ml/100 g body weight). To prepare the unilateral CCI, the right sciatic nerve was exposed at mid-thigh level by blunt dissection just proximal to its trifurcation, and three ligatures (3-0 sutures; Johnson&Johnson, Ethicon, Inc., Belgium) were applied to reduce the nerve diameter by one-third. Animals who underwent CCI were left to survive for 1 (n = 21), 3 (n = 21), 7 (n = 21), or 14 (n = 21) days. The naïve control group consisted of 21 intact rats. Sham-operated rats (sham group; n = 54) had the right sciatic nerves exposed only, without lesion, and were allowed to survive for 1 (n = 21), 3 (n = 21), and 14 (n = 12) days. The assessors of the experimental groups were blinded to treatment (CCI versus sham) for all types of measurement.

Behavioral tests

Withdrawal thresholds for mechanical hyperalgesia and latencies for thermal hyperalgesia were measured in both ipsilateral and contralateral hind and forepaws by dynamic plantar esthesiometer and plantar test (Ugo Basile, Italy), respectively. Rats were first acclimated in clear Plexiglas boxes for 30 minutes prior to testing. The paws were tested alternately with a 5 minute interval between tests. Six threshold and six latency measurements were taken for each paw during each test session 1 day before and 1, 3, 7, and 14 days after operation.
For thermal hyperalgesia, withdrawal time was measured and the intensity radiance was set to a value of 50. Data are expressed as mean ± SD of withdrawal thresholds (grams) and withdrawal latencies (seconds) for mechanical and thermal hyperalgesia, respectively.

Immunohistochemical staining

Three naive rats and three rats for each period of survival from CCI (1, 3, 7, and 14 days) and sham operation (1, 3, and 14 days) were deeply anesthetized with a lethal dose of sodium pentobarbital (70 mg/kg body weight, IP) and perfused transcardially with 500 ml phosphate-buffered saline (PBS: 10 mmol/l sodium phosphate buffer, pH 7.4, containing 0.15 mol/l NaCl) followed by 500 ml of Zamboni’s fixative [23]. The L4-L5 and C7-C8 DRG from both sides were detected within their intervertebral foramina after total laminectomy and foraminotomy. The DRG were removed, immersed separately in Zamboni’s fixative at 4°C overnight, and then collected separately into samples of ipsilateral lumbar (L-DRGi), contralateral lumbar (L-DRGc), ipsilateral cervical (C-DRGi) and contralateral cervical (C-DRGc) DRG for each period of survival and each group of rats (naive, sham, and CCI). The samples were washed in 20% phosphate-buffered sucrose for 12 hours. Pairs of ipsilateral and contralateral DRG (C7-C8 or L4-L5 segments) were embedded in optimal cutting temperature compound (Tissue-Tek® OCT compound; Miles, Elkhart, IN USA) and cut together. Serial longitudinal cryostat sections (12 μm) through the DRG were mounted on chrome-alum coated slides, and processed for indirect immunohistochemical staining, which was performed simultaneously for lumbar and cervical segments.
Interleukin-6 and interleukin-6 receptor immunofluorescence. Sections were washed with PBS containing 0.05% Tween 20 (PBS-T) and 1% bovine serum albumin (BSA) for 10 minutes, treated with 5% normal donkey serum in PBS-T for 30 minutes, then incubated with 25 μl of rabbit polyclonal antibodies against IL-6 (1:500; Invitrogen Inc., Camarillo, CA, USA) or IL-6R (1:200; Santa Cruz Biotechnology, Santa Cruz, CA, USA) in a humid chamber at room temperature (21 to 23°C) for 12 hours. The immunohistochemical reaction was visualized by treatment with tetramethyl rhodamine isothiocyanate (TRITC)-conjugated and affinity-purified donkey anti-rabbit secondary antibody (1:100; Millipor, Billerica, MA, USA) for 90 minutes at room temperature. The control sections were incubated without the primary antibody or with the primary antibody saturated by recombinant rat IL-6 protein (Invitrogen). Sections were stained with Hoechst 33342 to detect positions of the cell nuclei, mounted in aqueous mounting medium (Vectashield; Vector Laboratories Inc., Burlingame, CA, USA) and analyzed using an epifluorescence microscope (DMLB; Leica Microsystems GmbH, Wetzlar Germany) equipped with a camera (DFC-480; Leica Microsystems) and a stabilized power supply for the lamp housing. The same immunostaining pattern for IL-6 and interleukin-6 receptor (IL-6R) was seen in the DRG of L4-L5 and of C7-C8 spinal-cord segments removed from the same side of naive, CCI and sham rats. Therefore, the results are described for the lumbar or cervical DRG of the ipsilateral or contralateral side.
Double immunostaining. Some of the sections taken through ipsilateral lumbar DRG from CCI rats surviving for 3 or 7 days were double-stained. After incubation with rabbit polyclonal anti–IL-6 antibody for 12 hours and intensive washing, the sections were covered with mouse monoclonal anti-glutamine synthase (anti-GS; 1:500; LS-C23895; LifeSpan BioSciences, Inc., Seattle, WA, USA), anti-CD68 (ED-1; 1:100; MCA341R) or anti-T-cell receptor (anti-TCR; 1:50; MCA453G) (both Serotec, Düsseldorf, Germany) antibodies and incubated for 4 hours. A mixture (1:1) of affinity-purified TRITC-conjugated donkey anti-rabbit and fluorescein isothiocyanate (FITC)-conjugated donkey anti-mouse secondary antibodies (Millipor, Billerica, MA, USA) was applied at a final dilution of 1:100 for 90 minutes at room temperature.
To visualize colocalization of IL-6 and activating transcription factor (ATF-3) indicating neuronal bodies with injured axons [24], the sections were incubated with monoclonal anti-IL-6 antibody (1:100; ARC0962; BioSource, Camarillo, CA, USA) and then rabbit polyclonal anti-ATF3 (1:200; sc188; Santa Cruz Biotechnology, Santa Cruz, CA, USA). After intensive washing, affinity-purified TRITC-conjugated donkey anti-mouse and FITC-conjugated donkey anti-rabbit secondary antibodies (Millipor, Billerica, MA, USA) were applied at a final dilution of 1:100 for 90 minutes at room temperature.
Sections incubated with rabbit polyclonal anti-IL-6R antibody for 12 hours were then treated with mouse monoclonal anti-GS for 4 hours. To visualize colocalization, the sections were incubated with TRITC-conjugated donkey anti-rabbit and FITC-conjugated donkey anti-mouse secondary antibodies at room temperature for 90 minutes.

ELISA

Six naive rats and, CCI rats surviving for 1 (n = 6), 3 (n = 6), 7 (n = 6), and 14 (n = 6) days, and sham rats surviving for 1 (n = 6), 3 (n = 6), and 14 (n = 6) days were killed by CO2 inhalation. Blood samples were obtained by intracardiac puncture, and collected into tubes containing heparin and protease inhibitor cocktail (LaRoche, Basel, Switzerland). Plasma was immediately separated by low-speed centrifugation (2,500 g for 12 minutes).
Both ipsilateral and contralateral L4-L5 and C6-C7 DRG were removed and immediately collected in ice-cold PBS-T containing protease inhibitor cocktail (LaRoche, Basel, Switzerland). The DRG samples were divided into distinct groups of lumbar and cervical naive DRG (C-DRGn, L-DRGn) and ipsilateral and contralateral lumbar and cervical DRG (L-DRGi, L-DRGc, C-DRGi, and C-DRGc) taken from both CCI and sham rats for each period of survival. The DRG samples were homogenized in ice-cold PBS-T and separated by centrifugation (12,500 g for 12 minutes) to obtain extract proteins.
The tissue supernatant and plasma samples were stored at −60°C until analyzed. The total protein concentration was measured by spectrophotometer (Nanodrop ND-1000; Thermo Fisher Scientific Inc., Rockford, IL, USA) and the level of IL-6 protein was assessed by ELISA using a commercial kit with a sensitivity of 5 pg/ml (BioSource, Camarillo, CA, USA) in accordance with the manufacturer’s instructions. Each sample was measured five times using a microplate reader (SUNRISE Basic; Tecan, Salzburg, Austria) and data were standardized as pg of IL-6 protein to 100 μg of total protein. The IL-6 protein levels were normalized to baseline values of DRG and plasma from naive rats, which were set as 1, and final data are expressed as mean ± SD.

Western blotting analysis

Naive rats (n = 6), CCI rats surviving for 1 (n = 6), 3 (n = 6), 7 (n = 6), and 14 (n = 6) days, and sham rats surviving for 1 (n = 6) or 3 (n = 6) days were deeply anesthetized with a lethal dose of sodium pentobarbital (70 mg/kg body weight, IP.). DRG of both sides were then detected within their intervertebral foramina after total laminectomy and foraminotomy. Whole DRG were extracted under aseptic conditions from L4-L5 and C7-C8 levels, and classified as ipsilateral lumbar (L-DRGi), contralateral lumbar (L-DRGc), ipsilateral cervical (C-DRGi), and contralateral cervical (C-DRGc) DRG for each period of survival and each group of rats (naive, CCI, and sham). These were fast-frozen in liquid nitrogen, then stored at −65°C until the time of analysis. For triplicate western blotting analysis, samples of DRG were collected from two rats in each group. The samples were homogenized in PBS containing 0.1% Triton X-100 and protease inhibitors (LaRoche) and separated by centrifugation at 10,000 g for 5 minutes at 4°C. The total protein concentration was measured in the tissue supernatant (Nanodrop ND-1000; Thermo Fisher Scientific) and normalized to the same levels. Proteins were separated by SDS-polyacrylamide gel electrophoresis [25] and transferred to nitrocellulose membranes by electroblotting (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Blots were blocked by 1% BSA in PBS-T (3.2 mmol/l Na2HPO4, 0.5 mmol/l KH2PO4, 1.3 mmol/l KCl, 135 mmol/l NaCl, 0.05% Tween 20, pH 7.4) for 1 hours and incubated with anti-IL-6 polyclonal antibody (1:500; ARC0062; Biosource) overnight. Blots were washed in PBS-T and incubated with peroxidase-conjugated anti-rabbit IgG (1:1000; Sigma-Aldrich, St Louis, MO, USA) for 1 hour at room temperature. Equal loading of proteins was confirmed by α-tubulin staining. Protein bands were visualized using a chemiluminescence detection kit (ECL kit; Amersham Biosciences Inc., Piscataway, NJ, USA) on a chemiluminometer reader (LAS-3000; Bouchet Biotech) and analyzed using densitometry image software. After normalization to tubulin, IL-6 protein data were expressed as mean fold change relative to naive DRG, which was set as 1.

In situ hybridization

For DRG tissue harvesting, three naive rats and three rats for each period of survival were deeply anesthetized with a lethal dose of sodium pentobarbital (70 mg/kg body weight, IP), and perfused transcardially with 500 ml PBS containing 0.1% diethylpyrocarbonate (DEPC), followed by 500 ml of 4% paraformaldehyde with 0.1% DEPC. The DRG samples were washed in 20% phosphate-buffered sucrose for 12 hours and embedded in OCT compound (Tissue-Tek®; Miles). Serial longitudinal sections 12 μm thick through the DRG were cut on a cryostat and then mounted on chrome-alum coated slides.
To localize the IL-6 gene transcript, in situ hybridization was performed in accordance with the protocol of Harnicarova and coworkers [26]. We used two 50-mer oligoprobes (VBC-Biotech, Vienna, Austria) synthesized for the target IL-6 gene transcript (Table 1). Digoxigenin (DIG)-dT was used for probe labeling. All solutions used in this procedure were prepared in double-distilled water treated with DEPC. DIG was detected with a commercial kit (DIG Colorimetric Nucleic Acid Detection Kit; LaRoche). The sections were mounted in aqueous mounting medium (Vectashield; Vector Laboratories) and analyzed using a microscope (DMLB; Leica Microsystems) equipped with a camera (DFC-480; Leica Microsystems). The control sections that had been incubated without the DIG-oligonucleotide probes displayed no color staining.
Table 1
Probes used for in situ hybridization.
Probe
Sequence (5′→3′)
1
CGCTGTTCATACAAT*CAGAATTGCCAT*TGCACAACTCT*TTTCTCATTTCC
2
TCAAGTGCTTTCAAGAT*GAGTTGGATGGTCTTGGT*CCTTAGCCACTCCTTC
Asterisks indicate position of digoxigenin labeling.

Image analysis

The neuronal diameter, immunofluorescence intensity (brightness), and density of mRNA staining were assessed using an image analysis system (LUCIA-G; Laboratory Imaging Ltd., Prague, Czech Republic) in accordance with our previously published protocol [27]. Briefly, stained structures were detected for measurement after subtraction of background by the interactive thresholding technique (HSI: hue, saturation, and intensity) implemented in the image analysis (LUCIA) software, then transformed to binary mode. The binary foreground was monitored at every step of thresholding and manually edited if needed. The original color image was converted to gray and overlaid with the binary map. At least 200 neuronal profiles containing nuclei were measured for short (1 and 3 days) and late (7 and 14 days) periods of survival. The diameters of the DRG neurons were calculated from areas of neuronal profiles in sections for immunofluorescence and in situ hybridization, and the sizes of the DRG neurons were categorized as small (<25 μm), medium (25–40 μm), or large (>40 μm). The immunofluorescence and mRNA staining intensities were normalized to values of naive DRG and expressed as mean fold increase of intensity ± SD.

Statistical analyses

Behavioral data were evaluated using Kruskal-Wallis one-way analysis with Bonferroni post hoc test and P<0.05 was considered significant. To verify differences in ELISA, a Bonferroni-corrected one-way ANOVA for repeated measures was run, with P<0.05 as the level of significant difference between tested samples. Data for naive and CCI rats for intensity of IL-6 and IL-6R immunostaining and density of IL-6 mRNA were tested using the Mann-Whitney U-test (P<0.05). All statistical analyses were performed using STATISTICA software (version 9.0; StatSoft Inc., Tulsa, OK, USA).

Results

Behavioral tests

All rats with CCI of the sciatic nerve displayed decreased thresholds of mechanical hyperalgesia and withdrawal latencies of thermal hyperalgesia restricted to the hind paws ipsilateral to nerve ligatures as signs of neuropathic pain. There were no significant changes in withdrawal threshold for mechanical and thermal hyperalgesia in the contralateral hind paws compared with the results 1 day before operation. Thresholds of mechanical and thermal hyperalgesia did not significantly change in either the ipsilateral or contralateral fore paws (Figure 1A).
Both ipsilateral and contralateral hind paws of sham rats exhibited a decrease in thresholds of mechanical hyperalgesia between days 3 and 14, but there was no statistical significance when compared with 1 day before surgical treatment. By contrast, significantly decreased withdrawal latencies of thermal hyperalgesia were found 1 and 3 days after the sham operation. This bilateral hypersensitivity for thermal stimuli was present in both fore and hind paws, and normalized during the following days of survival after the sham operation (Figure 1B).

Immunohistochemical staining

Interleukin-6 immunofluorescence

Sections of lumbar and cervical DRG from naive rats showed weak immunofluorescence staining for IL-6 protein in small and medium-sized neuronal bodies and in SGC enveloping large neuronal bodies (Figure 2A,B). An increased intensity of IL-6 immunofluorescence was induced in all neuronal types of both ipsilateral and contralateral DRG removed from lumbar and cervical levels of all rats surviving up to 14 days after unilateral CCI of the sciatic nerve. A distinct increase in immunofluorescence intensity was particularly apparent in large neuronal bodies (>40 μm) compared with DRG removed from naive rats. The sections of lumbar and cervical DRG obtained from sham rats displayed a similar pattern and intensity of IL-6 immunofluorescence staining in the neuronal bodies to those removed from naive rats. In contrast to naive DRG, however, moderate intensity of IL-6 immunostaining was seen in SGC. Representative immunostaining for IL-6 in sections of lumbar and cervical DRG removed from both sides of rats surviving 3 days after CCI and sham operations is shown in Figure 2.
The control sections of DRG from CCI rats incubated with omission of the primary antibody or with the primary antibody saturated with protein were free of immunofluorescence staining (data not shown).
Immunofluorescence for ATF-3, indicating neuronal bodies with injured axons [24], was found in neuronal nuclei of lumbar DRG ipsilateral to CCI but not in cervical DRG. Only a few neurons of contralateral lumbar DRG displayed ATF-3 immunostaining. The increased IL-6 immunoreactivity in ipsilateral lumbar DRG was present not only in ATF-3-positive but also in ATF-3-free neurons (Figure 3A–C). Double immunostaining also confirmed an increase in IL-6 protein in SGC (Figure 3D–F), and provided evidence of a contribution of ED-1+ macrophages and TCR+ cells to the IL-6 level in DRG removed from rats subjected to CCI (Figure 4).
Compared with naive DRG, a significant increase in IL-6 immunofluorescence intensity was detected in large, medium, and small neuronal bodies of both lumbar and cervical DRG from rats surviving for short (3 days) and long (14 days) periods after CCI treatment. The highest increase in IL-6 intensity was found in large neurons of lumbar DRG ipsilateral to CCI, and this level was significantly higher than the contralateral counterpart. However, the large neurons of cervical DRG and the medium and small neurons of both cervical and lumbar DRG displayed increased immunofluorescence intensity without significant differences when the ipsilateral and contralateral sides were compared in each period of survival. The IL-6 intensity of large neurons of ipsilateral lumbar DRG was significantly reduced when we compared short and long periods of survival, whereas a bilateral decrease was found in medium neurons for the same comparison. Small neurons in lumbar DRG and all types of neurons in cervical DRG did not display significant changes when short and long periods were compared (Figure 5).

Interleukin-6 receptor immunofluorescence

Sections of naive lumbar and cervical DRG displayed IL-6R immunofluorescence of moderate intensity in medium and small neurons, whereas large neuronal bodies had very weak or no immunostaining. Compared with naive DRG, there was a significant increase in IL-6R immunofluorescence bilaterally in all neurons of lumbar DRG from CCI rats for short (3 days) and long (14 days) periods of survival. Distinct immunofluorescence was also present in SGC, surrounding mainly large neurons of ipsilateral lumbar DRG (Figure 6). This was confirmed by double immunostaining for IL-6R and with GS used as a marker for SGC (Figure 7). There was a significantly increased intensity of IL-6R immunostaining in the neuronal bodies of all diameters in both lumbar and cervical DRG of CCI rats after 3 and 14 days (Figure 8). A significantly higher level of IL-6R immunofluorescence was found in the large neurons of lumbar DRG from the ipsilateral compared with the contralateral side, but no significant differences were found between the ipsilateral and contralateral medium and small neurons 3 days after CCI. The level of IL-6R immunofluorescence was significantly decreased in all types of neurons 14 days after CCI.
Neuronal bodies of all diameters also displayed significant increases in IL-6R immunofluorescence in the cervical DRG of the ipsilateral side compared with naive neuronal bodies. This increased immunostaining in ipsilateral cervical DRG was found at 3 and 14 days from CCI. The level was significantly higher in large and medium-sized neurons in the ipsilateral compared with the contralateral cervical DRG. By contrast, the small neurons displayed bilateral elevation of IL-6R immunostaining after both short and long periods of survival.

ELISA assessment of interleukin-6 protein in dorsal root ganglia and plasma

The baseline level of IL-6 protein was significantly higher in cervical (124.9 ± 8.1 pg/0.1 mg total protein) than lumbar (75.1 ± 6.0 pg/0.1 mg total protein) DRG of the naive rats. Thus, the differences in DRG of different spinal levels should be borne in mind when comparing results of operated animals. Therefore, the IL-6 protein levels were normalized to the baseline values of the naive rats to compare IL-6 alteration in DRG and plasma of CCI and sham rats.
Compared with DRG of naive rats, increased levels of IL-6 protein were found bilaterally in both lumbar and cervical DRG from 1 to 14 days after unilateral CCI of the sciatic nerve. The levels of IL-6 protein peaked on day 3, when they were 10 and more than 5 times higher in ipsilateral and contralateral lumbar DRG, respectively. These levels declined afterwards, but remained higher than those of naive DRG (Figure 9).
Significant increases in IL-6 levels were found in ipsilateral lumbar and cervical DRG from sham rats surviving for 1, 3, and 14 days. The corresponding contralateral DRG did not display significant elevation of IL-6 protein compared with naive DRG (Figure 9).
The IL-6 protein levels increased approximately 1.5 and 2 times in the plasma of CCI and sham rats surviving for 1 and 3 days, respectively. Levels of plasma IL-6 protein then normalized during the time to the next survival time points (Figure 10).

Western blot analysis

Western blot analysis showed bilateral increases in IL-6 protein levels in both lumbar (Figure 11A) and cervical DRG (Figure 11B) of CCI rats compared with DRG of naive DRG. Densitometry of western blots for total L4-L5 DRG fractions showed a significant bilateral increase in IL-6 protein at 1, 3, 7, and 14 days after surgery. At 1 and 3 days after CCI, the levels of IL-6 protein were increased bilaterally by about 6 (L4-L5 DRG) and 5 (C7-C8 DRG) times in comparison with DRG of the naive rats. The levels of IL-6 protein remained similarly increased, by more than 3 times, in DRG of both lumbar and cervical segments at postoperative days 7 and 14.
Compared with naive rats, sham rats had higher levels of IL-6 protein in both lumbar and cervical DRG removed 1 day after the sham operation. The IL-6 protein levels in DRG from sham rats surviving for 3 days were similar to those of naive rats.

In situ hybridization

Neuronal bodies and their SGC displayed no or very weak signal for IL-6 mRNA in sections of lumbar and cervical DRG removed from naive rats. Faint staining for IL-6 mRNA was detected only in the blood vessels of naive DRG (Figure 12A, Figure 13A). Unilateral CCI of the sciatic nerve induced conspicuous bilateral elevation of staining for IL-6 mRNA in both lumbar and cervical DRG 1, 3, 7, and 14 days after operation. The patterns and intensities of staining for IL-6 mRNA were very similar in lumbar and cervical DRG sections for 1 and 3 days and for 7 and 14 days of survival, and hence representative illustrations from 3 and 14 days are shown (Figure 12B–E, Figure 13B,C). In comparison with naive DRG, an increased intensity for IL-6 mRNA staining was seen, particularly in neuronal bodies of all sizes and in the SGC surrounding the large neurons. No detectable signal for IL-6 mRNA was found in cells of the DRG capsule, but there was staining lining the Schwann cells of nerve fibers inside the DRG.
When the DRG sections were treated for in situ hybridization under the same conditions, the density of IL-6 mRNA staining was higher in sections of ipsilateral than contralateral lumbar DRG for short (1 and 3 days) and long (7 and 14 days) periods of survival (Figure 14). The intense staining for IL-6 mRNA in neuronal bodies did not usually allow staining to be distinguished in the thin layer of the SGC envelope. This was possible only in some SGC surrounding the large neuronal bodies and mainly in DRG from sham rats (Figure 12F–I; Figure 13E).
Compared with lumbar DRG, the sections of cervical DRG from CCI rats had weaker staining for IL-6 mRNA in neuronal bodies, and therefore the staining of SGC was highlighted (Figures 13B,C). The density of IL-6 mRNA staining was slightly higher in medium and small neurons for short (1 and 3 days) than for long (7 and 14 days) periods of survival. No marked differences in IL-6 mRNA staining were seen between ipsilateral and contralateral cervical DRG (Figure 14).
In comparison with naive DRG, stronger staining for IL-6 mRNA was seen in SGC and small neuronal bodies of both ipsilateral and contralateral lumbar DRG removed after 3 days from sham rats. Increased staining intensity was seen in SGC and blood vessels but no neuronal staining was found in lumbar DRG of rats surviving 14 days after sham operation (Figure 12F–I). Although at 3 days after sham operation, the cervical DRG on both sides displayed increased staining for IL-6 mRNA in blood vessels only, a weak signal was found in SGC when DRG were removed 14 days after sham operation (Figure 13D–E).

Discussion

IL-6 is a multifunctional cytokine whose increased level in the nervous system is rapidly and strongly induced by injury and by pathological and inflammatory stimuli [28]. A very low level of IL-6 was reported in the peripheral nervous system of intact mature animals, but its increase is induced distal to the site of sciatic nerve injury [29, 30] and in the large and medium lumbar DRG neurons [15, 31]. A weaker IL-6 induction was found in a CCI model of neuropathic pain, but it persisted for a longer time than in a nerve transection model. IL-6 induction in the CCI model was shown to correlate well with the duration of hypersensitivity [16]. It was reported that intrathecally administered human recombinant IL-6 elicited touch-evoked hyperalgesia in normal rats [32] and an injection of IL-6 into the rat hind paw induced dose-dependent mechanical hyperalgesia [33]. These findings suggest that IL-6 may be centrally involved in the cascade of events leading to the development of neuropathic pain. However, in addition to cytokines, upregulation of other immune mediators such as chemokines may also increase excitation of the DRG neurons in reaction to nerve injury [34, 35].

Cellular localization of interleukin-6 protein and mRNA in dorsal root ganglia

In agreement with previous papers [30, 36], our results for immunohistochemical staining and in situ hybridization show that the primary sensory neurons are significant sources for enhanced IL-6 in DRG after CCI of the sciatic nerve. As indicated by ATF-3 staining, increased IL-6 expression was not limited to neurons with axonal injury but was also present in all neurons of DRG. In particular, significant enhancement of IL-6 immunostaining was clearly visible in large neuronal bodies, with greater enhancement in lumbar DRG ipsilateral to CCI than in contralateral DRG. The neuronal bodies of all sizes in cervical DRG and the medium and small neurons of lumbar DRG had increased IL-6 immunostaining but without significant differences between ipsilateral and contralateral sides. Double immunostaining showed that SGC, macrophages, and T cells may also contribute to the enhanced level of IL-6 protein in DRG.
Very little or no IL-6 protein or mRNA were detected in DRG neurons of sham rats, indicating that intraneuronal occurrence of IL-6 protein and its synthesis is induced by unilateral CCI of the sciatic nerve. Intraneuronal IL-6 mRNA and protein have been detected in DRG after various types of nerve manipulation [15, 31, 37]. In contrast to previous reports of induction of IL-6 mRNA only in the medium-sized and large lumbar DRG neurons [15, 31], we found distinct increases in signal for IL-6 mRNA in DRG neurons of all sizes. This discrepancy is possibly due to the different in situ hybridization methods used, with radiographic probes being used by Murphy and coworkers (35S-labeled or 33P-labeled oligonucleotides) but a non-radioactive method (DIG-labeled probes) in our experiments. An in situ hybridization method with DIG-labeled probes can be used successfully to detect mRNAs in frozen sections with a sensitivity equal to or better than that of a radioactive method but with a much higher cellular resolution [38]. Therefore, an in situ hybridization method with DIG-labeled probes was used successfully in our experiments to detect IL-6 mRNA in neuronal bodies and their SGC in frozen DRG sections removed from rats.
It is well-known that SGC are activated by nerve injury, and they may also play a role in the development of pathological pain [39]. Compared with naive DRG, there was higher intensity of immunofluorescence for IL-6 and of staining for IL-6 mRNA in SGC of lumbar and cervical DRG of both sides after CCI or sham operation. However, it was difficult to distinguish unequivocally between IL-6 mRNA staining in neuronal bodies and their SGC of lumbar DRG, because a similar staining intensity was present in both cell types. Detection of IL-6 mRNA in SGC was easier in sections of cervical DRG when the staining density of neuronal bodies was lower than in lumbar DRG. Moreover, in contrast to neurons, SGC of both lumbar and cervical DRG removed from sham rats displayed increased staining for IL-6 mRNA, thus indicating that their activation and resultant synthesis of IL-6 may occurre only in response to tissue damage during surgical treatment. These results of activated SGC in DRG of sham rats correspond with findings of SGC proliferation and activation in response to scarification or incision of the skin [40, 41].
IL-6 acts by binding to IL-6R and activating the gpl30 transducer chain. Although IL-6R expression is limited in cells of the nervous system [42], we found bilateral increases in IL-6R immunostaining in all neurons of both lumbar and cervical DRG from CCI rats. However, only SGC of lumbar DRG ipsilateral to CCI of the sciatic nerve displayed distinct immunostaining for IL-6R. This indicates some small differences of IL-6 action in DRG directly associated and not associated with injured nerve [43].

Bilateral expression of interleukin-6 protein and mRNA in dorsal root ganglia after unilateral chronic constriction injury of the sciatic nerve

Bilateral expression of IL-6 protein and mRNA in DRG was not unexpected, because there is a growing body of evidence that unilateral nerve damage results in bilateral changes in neurochemical and electrophysiological parameters in DRG [4446], including cytokines [18, 37, 47, 48]. It has been generally accepted that contralateral responses to unilateral nerve injury are usually qualitatively similar but smaller in magnitude and have a briefer time course compared with ipsilateral changes [49]. However, our results showed that levels of IL-6 protein and mRNA in the contralateral DRG paralleled those of the ipsilateral DRG, not only in homonymous but also in heteronymous spinal cord segments with injured nerve. Bilateral upregulation of IL-6 in lumbar and cervical DRG after unilateral nerve injury is comparable with the expression of tumor necrosis factor (TNF)-α and IL-10 [50, 51]. This indicates that cytokine upregulations in DRG that are associated or not associated with damaged nerve are induced by similar mechanisms.
The original CCI method using chromic gut [52] is a widely applied experimental model that induces characteristic signs and symptoms of neuropathic pain found in humans. However, because chromic gut itself induces local inflammatory reaction, this original CCI model of neuropathic pain is not suitable for distinguishing neuroinflammatory reactions induced by a thread material and/or Wallerian degeneration of injured axons [53, 54]. Therefore, we prepared CCI of the sciatic nerve in our experimental rats using 3-0 sterilized suture (Ethicon) under aseptic conditions. Thus, the bilateral changes in IL-6 protein and mRNA in both lumbar and cervical DRG presented here were largely induced by partial traumatic nerve injury accompanied by neuroinflammatory response of Wallerian degeneration.
Possible mechanisms of contralateral signaling were reviewed by Koltzenburg and coworkers [49], but the underlying molecular mechanisms and neuroanatomical pathways linked with bilateral DRG responses to unilateral peripheral nerve injury remain largely unknown. Two main types of stimuli may be involved in inducing bilateral changes in IL-6 protein and mRNA in both lumbar and cervical DRG after unilateral CCI of the sciatic nerve. The first type of stimuli could be transferred by neuronal pathways, for example, through interneurons at the spinal cord and supraspinal levels [5557]. Moreover, there is a long ascending propriospinal system linking lumbar and cervical spinal-cord segments. The so-called long ascending propriospinal neurons are defined as interneurons whose somata are located in the lumbar spinal-cord segments and whose axons terminate in cervical segments. These neurons are in an anatomically appropriate position to participate in coordinating movements of hind and fore limbs [58]. Changes in neuronal activity may partially contribute to the induction of IL-6 expression in neurons [59], although other mechanisms of IL-6 regulation, for example by systemic factors, can be assumed because DRG do not contain a complete blood–nerve barrier [60].
These other possible mechanisms inducing IL-6 mRNA and protein in DRG are probably related to production of signaling molecules during Wallerian degeneration. Experimental findings have suggested that IL-6 is induced in DRG by an injury factor arising from the nerve stump rather than by interruption of retrograde axonal transport of signal molecules from target tissues or distal nerve segments [15, 16]. The results from the current study implicate Wallerian degeneration as one possible source of the factors inducing bilateral increases in IL-6 mRNA and protein in both lumbar and cervical DRG after unilateral CCI performed under aseptic conditions. The type of signal molecules produced by Wallerian degeneration can pass through an interrupted blood–nerve barrier [61, 62], thus allowing diffusion of circulating signal molecules into the microenvironment of the DRG not associated with the injured nerve. Several candidate molecules have been suggested for signaling from damaged nerve, including ATP, glutamate, complement or prostaglandin E2 [6366]. Some of these signal molecules are probably produced by damaged tissue during surgical treatment, as was indicated by induction of IL-6 mRNA and protein in SGC of DRG removed from sham rats.
ATP, which is suggested to be one of the first mediators of tissue damage, acts through ionotropic P2X receptors and metabotropic P2Y receptors [67]. It has been shown that the P2X3 receptor of DRG neurons and the P2X7 receptor expressed in SGC are two major purinergic receptors participating in neuron–SGC communication. The bodies of excited DRG neurons release ATP and activate P2X7 receptor, which is expressed only in SGCs [6870]. Although it is noteworthy that P2X7 receptor is associated with inflammatory reactions of SGC in order to induce their sensitivity to ATP, and that this may significantly contribute to neuropathic pain [71], it has been shown that activation of P2X3 contributing to mechanical hyperalgesia does not depend on pro-inflammatory cytokines, including IL-6 [72]. However, the P2X7 receptor is upregulated in human DRG and injured nerves obtained from patients with chronic neuropathic pain. It has been reported that P2X7 receptor knockout animals did not display mechanical or thermal hyperalgesia although normal nociceptive processing was preserved [73]. However, involvement of the P2X7 receptor in the regulation of IL-6 is controversial. It was reported that ATP-induced IL-6 production is not mediated by P2X7 receptors [74], whereas others found markedly increased levels of IL-6 in inflamed hind paw of P2X7-/- mice [73], indicating that this receptor is involved in regulating IL-6 levels. This might be present in the case of SGC, which, in contrast to DRG neurons, express both P2X7 [73] and IL-6.

Serum level of interleukin-6 protein after chronic constriction injury of the sciatic nerve

There are some controversial results illustrating changes in plasma IL-6 after rat nerve injury and the involvement of these changes in neuropathic pain induction. It was reported that an increase in plasma IL-6 had no effect on pain [14], whereas other evidence suggested that such an increase had a hyperalgesic effect [33]. In our experiments, the plasma IL-6 protein was increased approximately 1.5 and 2 times in rats surviving 1 and 3 days from CCI and sham operation, respectively. By contrast, the increase in IL-6 protein in DRG was significantly higher (3 to 10 times), thus suggesting that plasma IL-6 did not contribute substantially to the increase of the cytokine in DRG. A decrease in IL-6 to the level seen in naive rats 7 and 14 days after CCI, when mechanical hyperalgesia and thermal hyperalgesia were measured, indicates that changes in plasma IL-6 induced by nerve injury do not correlate exactly with induction and maintenance of neuropathic pain.
In the context of neuropathic pain, it is important to mention that pro-inflammatory cytokines, of which IL-6 is the main one, also induce behavioral discomfort or ‘sickness’, not only in response to infection but also after nerve trauma [75, 76]. Although the level of IL-6 protein in blood plasma has specifically been shown to be correlated with infection-induced sickness [75], its level in neuropathic pain status is controversial [14, 33, 77]. The animals in our experiments were housed under specific pathogen-free conditions, and behavioral tests were measured under conditions that would minimize stress on the animals.
The level of plasma IL-6 in both CCI and sham rats was increased only at 1 and 3 days of survival, and normalized thereafter. A higher level of plasma IL-6 during the short period of survival corresponds with published findings [78], and reflects a post-operation reaction that is probably related to bilateral thermal hyperalgesia in both hind and fore paws of sham-operated rats.
Our behavioral tests are in accordance with published results [79] showing that sham surgery without nerve manipulation is sufficient to induce temporary hyperalgesia, as also measured by some other authors [80, 81]. Bilateral thermal hyperalgesia was identical in both hind and fore paws of sham rats in our experiments, thus indicating that effects of surgery treatment are adequate to alter mechanisms of sensory processing in both fore and hind paws. However, the same pattern of thermal hyperalgesia was not seen in sham versus CCI rats. To explain these findings, we believe that the central control structures may be differently activated after sham and CCI treatment of animals and thus would have resulted in their different behavioral reactions to heat stimuli. The sham operation probably did not activate central control mechanisms, as these are triggered by nerve injury [8284].

A possible functional involvement of interleukin-6 protein in dorsal root ganglia associated or not associated with injured nerve

IL-6 may be critically involved in the cascade of events after nerve injury leading to the development and maintenance of behaviors suggestive of neuropathic pain. Fundamental evidence as to a role of IL-6 in nociception and hyperalgesia has been found by direct injection of IL-6 into experimental animals [14, 32]. In addition, IL-6-/- mice showed reduced heat sensitivity in a hot-plate behavioral test [85]. Our results confirmed the increase in IL-6 mRNA and protein in DRG neurons ipsilateral to CCI for at least 14 days when hypersensitivity was apparent in the ipsilateral hind paws [16]. This indicates that continuous upregulation of IL-6 in DRG associated with injured nerve is linked to induction of hyperalgesia. Different animal neuropathic pain models have been established to cover the diverse etiology and consequently the diverse clinical manifestation of neuropathic pain, with high validation and reproducibility. However, they have shortcomings and limitations that should be taken into consideration. Principally, the measured alterations in cutaneous sensory thresholds might be responses to nerve injury rather than integrated pain-related behavior reactions. Moreover, because for ethical reasons experimental animals usually survive only days or weeks, the clinical aspects of neuropathic pain are measured in years [86]. Thus, animal models are relevant predominantly for testing of induction of neuropathic pain, and have limited value for simulating chronic hypersensitivity changes in patients.
Our results from immunohistochemistry, western blotting, and in situ hybridization unequivocally showed an unexpected IL-6 response in contralateral lumbar and cervical DRG on both sides, which did not coincide with behavioral signs of hypersensitivity in corresponding paw skin. A similar pattern of expression was found for TNF-α and IL-10 [50, 51] suggesting that these changes in DRG occurring after unilateral CCI may reflect a general neuroinflammatory reaction of the nervous system to injury. Involvement of cytokines upregulated in remote DRG with neuropathic pain induction is still unknown, and other experiments are required to elucidate this.
Elevation of IL-6 in DRG that are not associated with injured nerves indicates a possible functional involvement of IL-6 other than that of neuropathic pain induction. Several lines of evidence have shown that IL-6 is implicated as a key component in the injury response of the nervous system. Consistent with our findings for IL-6, there is evidence that nerve growth factor mRNA is also increased bilaterally in lumbar and cervical DRG after unilateral crushing of the sciatic nerve [87]. Moreover, IL-6 plays a role in promoting neuronal survival [12] and axonal growth by DRG neurons [37]. Thus, IL-6 upregulation in the primary sensory neurons of DRG that are not associated with damaged nerve might be linked with conditioning of the intact neurons to regenerate their axons [88, 89].

Conclusion

In a rat model of neuropathic pain based on aseptic unilateral CCI of the sciatic nerve, we found that the contralateral L4-L5 DRG and the cervical DRG on both sides were not spared from IL-6 response to unilateral sciatic nerve injury even though these DRG were not directly linked with the damaged nerve. The increase in IL-6 protein and mRNA in the ipsilateral lumbar DRG only was related to (or at least coincided with) behavioral presentations of hyperalgesia in the corresponding limb.
Although these findings of increased IL-6 protein and mRNA in DRG associated with damaged nerve may support a role for IL-6 in developing neuropathic pain, the finding of IL-6 in DRG not associated with injured nerve argues against a direct coupling between IL-6 elevation in DRG and hypersensitivity. The results of our IL-6 study suggest that neuroinflammatory reaction of DRG to nerve injury is propagated alongside neuroaxis from lumbar to remote cervical segments. This phenomenon probably illustrates a general neuroinflammatory reaction of the nervous system to local nerve injury.

Acknowledgements

We thank Ms. Dana Kutějová, Ms. Marta Lněníčková, Mgr. Zuzana Veselková, Ms. Jitka Mikulášková, Ms. Stana Bartová and Mr. Lumír Trenčanský for their skilful technical assistance. This work was supported by the projects CEITEC - Central European Institute of Technology (CZ.1.05/1.1.00/02.0068) from European Regional Development Fund, SYLICA/286154 (FP7-REGPOT-2011-1) and grant AV0Z50040507/AV0Z50040702 (Academy of Sciences of the Czech Republic).
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

PD conceived, designed and coordinated the study and wrote the manuscript. VB conceived, designed, and coordinated the western blot and in situ hybridization analyses. IK and IS conceived, designed and carried out the experiments, and participated in acquiring and analyzing the presented data. All authors gave final approval to the version to be published.
Literatur
1.
Zurück zum Zitat Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, Hansson P, Hughes R, Nurmikko T, Serra J: Neuropathic pain - redefinition and a grading system for clinical and research purposes. Neurology 2008,70(18):1630–1635.CrossRefPubMed Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, Hansson P, Hughes R, Nurmikko T, Serra J: Neuropathic pain - redefinition and a grading system for clinical and research purposes. Neurology 2008,70(18):1630–1635.CrossRefPubMed
2.
Zurück zum Zitat Berger JV, Knaepen L, Janssen SPM, Jaken RJP, Marcus MAE, Joosten EAJ, Deumens R: Cellular and molecular insights into neuropathy-induced pain hypersensitivity for mechanism-based treatment approaches. Brain Res Rev 2011,67(1–2):282–310.CrossRefPubMed Berger JV, Knaepen L, Janssen SPM, Jaken RJP, Marcus MAE, Joosten EAJ, Deumens R: Cellular and molecular insights into neuropathy-induced pain hypersensitivity for mechanism-based treatment approaches. Brain Res Rev 2011,67(1–2):282–310.CrossRefPubMed
4.
Zurück zum Zitat Kirita T, Takebayashi T, Mizuno S, Takeuchi H, Kobayashi T, Fukao M, Yamashita T, Tohse N: Electrophysiologic changes in dorsal root ganglion neurons and behavioral changes in a lumbar radiculopathy model. Spine 2007,32(2):E65-E72.CrossRefPubMed Kirita T, Takebayashi T, Mizuno S, Takeuchi H, Kobayashi T, Fukao M, Yamashita T, Tohse N: Electrophysiologic changes in dorsal root ganglion neurons and behavioral changes in a lumbar radiculopathy model. Spine 2007,32(2):E65-E72.CrossRefPubMed
5.
Zurück zum Zitat Shinder V, Govrin-Lippmann R, Cohen S, Belenky M, Ilin P, Fried K, Wilkinson HA, Devor M: Structural basis of sympathetic-sensory coupling in rat and human dorsal root ganglia following peripheral nerve injury. J Neurocytol 1999,28(9):743–761.CrossRefPubMed Shinder V, Govrin-Lippmann R, Cohen S, Belenky M, Ilin P, Fried K, Wilkinson HA, Devor M: Structural basis of sympathetic-sensory coupling in rat and human dorsal root ganglia following peripheral nerve injury. J Neurocytol 1999,28(9):743–761.CrossRefPubMed
6.
Zurück zum Zitat Dubovy P, Tuckova L, Jancalek R, Svizenska I, Klusakova I: Increased invasion of ED-1 positive macrophages in both ipsi- and contralateral dorsal root ganglia following unilateral nerve injuries. Neurosci Lett 2007,427(2):88–93.CrossRefPubMed Dubovy P, Tuckova L, Jancalek R, Svizenska I, Klusakova I: Increased invasion of ED-1 positive macrophages in both ipsi- and contralateral dorsal root ganglia following unilateral nerve injuries. Neurosci Lett 2007,427(2):88–93.CrossRefPubMed
7.
Zurück zum Zitat Wang H, Sun H, Della Penna K, Benz RJ, Xu J, Gerhold DL, Holder DJ, Koblan KS: Chronic neuropathic pain is accompanied by global changes in gene expression and shares pathobiology with neurodegenerative diseases. Neuroscience 2002,114(3):529–546.CrossRefPubMed Wang H, Sun H, Della Penna K, Benz RJ, Xu J, Gerhold DL, Holder DJ, Koblan KS: Chronic neuropathic pain is accompanied by global changes in gene expression and shares pathobiology with neurodegenerative diseases. Neuroscience 2002,114(3):529–546.CrossRefPubMed
8.
Zurück zum Zitat Zhang Y, Wang YH, Zhang XH, Ge HY, Arendt-Nielsen L, Shao JM, Yue SW: Proteomic analysis of differential proteins related to the neuropathic pain and neuroprotection in the dorsal root ganglion following its chronic compression in rats. Exp Brain Res 2008,189(2):199–209.CrossRefPubMed Zhang Y, Wang YH, Zhang XH, Ge HY, Arendt-Nielsen L, Shao JM, Yue SW: Proteomic analysis of differential proteins related to the neuropathic pain and neuroprotection in the dorsal root ganglion following its chronic compression in rats. Exp Brain Res 2008,189(2):199–209.CrossRefPubMed
9.
Zurück zum Zitat DeLeo JA, Yezierski RP: The role of neuroinflammation and neuroimmune activation in persistent pain. Pain 2001,90(1–2):1–6.CrossRefPubMed DeLeo JA, Yezierski RP: The role of neuroinflammation and neuroimmune activation in persistent pain. Pain 2001,90(1–2):1–6.CrossRefPubMed
10.
Zurück zum Zitat Obreja O, Biasio W, Andratsch M, Lips KS, Rathee PK, Ludwig A, Rose-John S, Kress M: Fast modulation of heat-activated ionic current by proinflammatory interleukin 6 in rat sensory neurons. Brain 2005,128(7):1634–1641.CrossRefPubMed Obreja O, Biasio W, Andratsch M, Lips KS, Rathee PK, Ludwig A, Rose-John S, Kress M: Fast modulation of heat-activated ionic current by proinflammatory interleukin 6 in rat sensory neurons. Brain 2005,128(7):1634–1641.CrossRefPubMed
11.
Zurück zum Zitat Liu Z, Qiu YH, Li B, Ma SH, Peng YP: Neuroprotection of interleukin-6 against NMDA-induced apoptosis and its signal-transduction mechanisms. Neurotox Res 2011,19(3):484–495.CrossRefPubMed Liu Z, Qiu YH, Li B, Ma SH, Peng YP: Neuroprotection of interleukin-6 against NMDA-induced apoptosis and its signal-transduction mechanisms. Neurotox Res 2011,19(3):484–495.CrossRefPubMed
12.
Zurück zum Zitat Yang P, Wen HZ, Ou S, Cui J, Fan DH: IL-6 promotes regeneration and functional recovery after cortical spinal tract injury by reactivating intrinsic growth program of neurons and enhancing synapse formation. Exp Neurol 2012,236(1):19–27.CrossRefPubMed Yang P, Wen HZ, Ou S, Cui J, Fan DH: IL-6 promotes regeneration and functional recovery after cortical spinal tract injury by reactivating intrinsic growth program of neurons and enhancing synapse formation. Exp Neurol 2012,236(1):19–27.CrossRefPubMed
13.
Zurück zum Zitat Arruda JL, Sweitzer SA, Rutkowski MD, DeLeo JA: Intrathecal anti-IL-6 antibody and IgG attenuates peripheral nerve injury-induced mechanical allodynia in the rat: possible immune modulation in neuropathic pain. Brain Res 2000,879(1–2):216–225.CrossRefPubMed Arruda JL, Sweitzer SA, Rutkowski MD, DeLeo JA: Intrathecal anti-IL-6 antibody and IgG attenuates peripheral nerve injury-induced mechanical allodynia in the rat: possible immune modulation in neuropathic pain. Brain Res 2000,879(1–2):216–225.CrossRefPubMed
14.
Zurück zum Zitat Vissers KC, De Jongh RF, Hoffmann VL, Meert TF: Exogenous interleukin-6 increases cold allodynia in rats with a mononeuropathy. Cytokine 2005,30(4):154–159.CrossRefPubMed Vissers KC, De Jongh RF, Hoffmann VL, Meert TF: Exogenous interleukin-6 increases cold allodynia in rats with a mononeuropathy. Cytokine 2005,30(4):154–159.CrossRefPubMed
15.
Zurück zum Zitat Murphy PG, Borthwick LS, Johnston RS, Kuchel G, Richardson PM: Nature of the retrograde signal from injured nerves that induces interleukin-6 mRNA in neurons. J Neurosci 1999,19(10):3791–3800.PubMed Murphy PG, Borthwick LS, Johnston RS, Kuchel G, Richardson PM: Nature of the retrograde signal from injured nerves that induces interleukin-6 mRNA in neurons. J Neurosci 1999,19(10):3791–3800.PubMed
16.
Zurück zum Zitat Murphy PG, Ramer MS, Borthwick L, Gauldie J, Richardson PM, Bisby MA: Endogenous interleukin-6 contributes to hypersensitivity to cutaneous stimuli and changes in neuropeptides associated with chronic nerve constriction in mice. Eur J Neurosci 1999,11(7):2243–2253.CrossRefPubMed Murphy PG, Ramer MS, Borthwick L, Gauldie J, Richardson PM, Bisby MA: Endogenous interleukin-6 contributes to hypersensitivity to cutaneous stimuli and changes in neuropeptides associated with chronic nerve constriction in mice. Eur J Neurosci 1999,11(7):2243–2253.CrossRefPubMed
17.
Zurück zum Zitat Kleinschnitz C, Brinkhoff J, Sommer C, Stoll G: Contralateral cytokine gene induction after peripheral nerve lesions: Dependence on the mode of injury and NMDA receptor signaling. Mol Brain Res 2005,136(1–2):23–28.CrossRefPubMed Kleinschnitz C, Brinkhoff J, Sommer C, Stoll G: Contralateral cytokine gene induction after peripheral nerve lesions: Dependence on the mode of injury and NMDA receptor signaling. Mol Brain Res 2005,136(1–2):23–28.CrossRefPubMed
18.
Zurück zum Zitat Hatashita S, Sekiguchi M, Kobayashi H, Konno SI, Kikuchi SI: Contralateral neuropathic pain and neuropathology in dorsal root ganglion and spinal cord following hemilateral nerve injury in rats. Spine 2008,33(12):1344–1351.CrossRefPubMed Hatashita S, Sekiguchi M, Kobayashi H, Konno SI, Kikuchi SI: Contralateral neuropathic pain and neuropathology in dorsal root ganglion and spinal cord following hemilateral nerve injury in rats. Spine 2008,33(12):1344–1351.CrossRefPubMed
19.
Zurück zum Zitat Attal N, Jazat F, Kayser V, Guilbaud G: Further evidence for pain-related behaviors in a model of unilateral peripheral mononeuropathy. Pain 1990,41(2):235–251.CrossRefPubMed Attal N, Jazat F, Kayser V, Guilbaud G: Further evidence for pain-related behaviors in a model of unilateral peripheral mononeuropathy. Pain 1990,41(2):235–251.CrossRefPubMed
20.
Zurück zum Zitat Weissner W, Winterson BJ, Stuart-Tilley A, Devor M, Bove GM: Time course of substance P expression in dorsal root ganglia following complete spinal nerve transection. J Comp Neurol 2006,497(1):78–87.CrossRefPubMedPubMedCentral Weissner W, Winterson BJ, Stuart-Tilley A, Devor M, Bove GM: Time course of substance P expression in dorsal root ganglia following complete spinal nerve transection. J Comp Neurol 2006,497(1):78–87.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Komori N, Takemori N, Kim HK, Singh A, Hwang SH, Foreman RD, Chung K, Chung JM, Matsumoto H: Proteomics study of neuropathic and nonneuropathic dorsal root ganglia: altered protein regulation following segmental spinal nerve ligation injury. Physiol Genomics 2007,29(2):215–230.CrossRefPubMed Komori N, Takemori N, Kim HK, Singh A, Hwang SH, Foreman RD, Chung K, Chung JM, Matsumoto H: Proteomics study of neuropathic and nonneuropathic dorsal root ganglia: altered protein regulation following segmental spinal nerve ligation injury. Physiol Genomics 2007,29(2):215–230.CrossRefPubMed
22.
Zurück zum Zitat Zimmermann M: Ethical guidelines for investigations of experimental pain in conscious animals. Pain 1983,16(2):109–110.CrossRefPubMed Zimmermann M: Ethical guidelines for investigations of experimental pain in conscious animals. Pain 1983,16(2):109–110.CrossRefPubMed
23.
Zurück zum Zitat Zamboni L, DeMartin C: Buffered picric acid-formaldehyde - a new rapid fixative for electron microscopy. J Cell Biol 1967,35(2P2):A148. Zamboni L, DeMartin C: Buffered picric acid-formaldehyde - a new rapid fixative for electron microscopy. J Cell Biol 1967,35(2P2):A148.
24.
Zurück zum Zitat Tsujino H, Kondo E, Fukuoka T, Dai Y, Tokunaga A, Miki K, Yonenobu K, Ochi T, Noguchi K: Activating transcription factor 3 (ATF3) induction by axotomy in sensory and motoneurons: A novel neuronal marker of nerve injury. Mol Cell Neurosci 2000,15(2):170–182.CrossRefPubMed Tsujino H, Kondo E, Fukuoka T, Dai Y, Tokunaga A, Miki K, Yonenobu K, Ochi T, Noguchi K: Activating transcription factor 3 (ATF3) induction by axotomy in sensory and motoneurons: A novel neuronal marker of nerve injury. Mol Cell Neurosci 2000,15(2):170–182.CrossRefPubMed
25.
Zurück zum Zitat Brazda V, Klusakova I, Svizenska I, Veselkova Z, Dubovy P: Bilateral changes in IL-6 protein, but not in its receptor gp130, in rat dorsal root ganglia following sciatic nerve ligature. Cell Mol Neurobiol 2009,29(6–7):1053–1062.CrossRefPubMed Brazda V, Klusakova I, Svizenska I, Veselkova Z, Dubovy P: Bilateral changes in IL-6 protein, but not in its receptor gp130, in rat dorsal root ganglia following sciatic nerve ligature. Cell Mol Neurobiol 2009,29(6–7):1053–1062.CrossRefPubMed
26.
Zurück zum Zitat Harnicarova A, Kozubek S, Pachernik J, Krejci J, Bartova E: Distinct nuclear arrangement of active and inactive c-myc genes in control and differentiated colon carcinoma cells. Exp Cell Res 2006,312(20):4019–4035.CrossRefPubMed Harnicarova A, Kozubek S, Pachernik J, Krejci J, Bartova E: Distinct nuclear arrangement of active and inactive c-myc genes in control and differentiated colon carcinoma cells. Exp Cell Res 2006,312(20):4019–4035.CrossRefPubMed
27.
Zurück zum Zitat Dubovy P, Klusakova I, Svizenska I: A quantitative immunohistochemical study of the endoneurium in the rat dorsal and ventral spinal roots. Histochem Cell Biol 2002,117(6):473–480.CrossRefPubMed Dubovy P, Klusakova I, Svizenska I: A quantitative immunohistochemical study of the endoneurium in the rat dorsal and ventral spinal roots. Histochem Cell Biol 2002,117(6):473–480.CrossRefPubMed
28.
Zurück zum Zitat Spooren A, Kolmus K, Laureys G, Clinckers R, De Keyser J, Haegeman G, Gerlo S: Interleukin-6, a mental cytokine. Brain Res Rev 2011,67(1–2):157–183.CrossRefPubMed Spooren A, Kolmus K, Laureys G, Clinckers R, De Keyser J, Haegeman G, Gerlo S: Interleukin-6, a mental cytokine. Brain Res Rev 2011,67(1–2):157–183.CrossRefPubMed
29.
Zurück zum Zitat Bolin LM, Verity AN, Silver JE, Shooter EM, Abrams JS: Interleukin-6 production by Schwann cells and induction in sciatic nerve injury. J Neurochem 1995,64(2):850–858.CrossRefPubMed Bolin LM, Verity AN, Silver JE, Shooter EM, Abrams JS: Interleukin-6 production by Schwann cells and induction in sciatic nerve injury. J Neurochem 1995,64(2):850–858.CrossRefPubMed
30.
Zurück zum Zitat Bourde O, Kiefer R, Toyka KV, Hartung HP: Quantification of interleukin-6 mRNA in wallerian degeneration by competitive reverse transcription polymerase chain reaction. J Neuroimmunol 1996,69(1–2):135–140.PubMed Bourde O, Kiefer R, Toyka KV, Hartung HP: Quantification of interleukin-6 mRNA in wallerian degeneration by competitive reverse transcription polymerase chain reaction. J Neuroimmunol 1996,69(1–2):135–140.PubMed
31.
Zurück zum Zitat Murphy PG, Grondin J, Altares M, Richardson PM: Induction of interleukin-6 in axotomized sensory neurons. J Neurosci 1995,15(7):5130–5138.PubMed Murphy PG, Grondin J, Altares M, Richardson PM: Induction of interleukin-6 in axotomized sensory neurons. J Neurosci 1995,15(7):5130–5138.PubMed
32.
Zurück zum Zitat DeLeo JA, Colburn RW, Nichols M, Malhotra A: Interleukin-6-mediated hyperalgesia/allodynia and increased spinal IL-6 expression in a rat mononeuropathy model. J Interferon Cytokine Res 1996,16(9):695–700.CrossRefPubMed DeLeo JA, Colburn RW, Nichols M, Malhotra A: Interleukin-6-mediated hyperalgesia/allodynia and increased spinal IL-6 expression in a rat mononeuropathy model. J Interferon Cytokine Res 1996,16(9):695–700.CrossRefPubMed
33.
Zurück zum Zitat Cunha FQ, Poole S, Lorenzetti BB, Ferreira SH: The pivotal role of tumor-necrosis-factor-alpha in the development of inflammatory hyperalgesia. Brit J Pharmacol 1992,107(3):660–664.CrossRef Cunha FQ, Poole S, Lorenzetti BB, Ferreira SH: The pivotal role of tumor-necrosis-factor-alpha in the development of inflammatory hyperalgesia. Brit J Pharmacol 1992,107(3):660–664.CrossRef
34.
Zurück zum Zitat Kao DJ, Li AH, Chen JC, Luo RS, Chen YL, Lu JC, Wang HL: CC chemokine ligand 2 upregulates the current density and expression of TRPV1 channels and Na(v)1.8 sodium channels in dorsal root ganglion neurons. J Neuroinflamm 2012., 9: 10.1186/1742–2094–9-189 Kao DJ, Li AH, Chen JC, Luo RS, Chen YL, Lu JC, Wang HL: CC chemokine ligand 2 upregulates the current density and expression of TRPV1 channels and Na(v)1.8 sodium channels in dorsal root ganglion neurons. J Neuroinflamm 2012., 9: 10.1186/1742–2094–9-189
35.
Zurück zum Zitat Holmes FE, Arnott N, Vanderplank P, Kerr NCH, Longbrake EE, Popovich PG, Imai T, Combadiere C, Murphy PM, Wynick D: Intra-neural administration of fractalkine attenuates neuropathic pain-related behaviour. J Neurochem 2008,106(2):640–649.CrossRefPubMedPubMedCentral Holmes FE, Arnott N, Vanderplank P, Kerr NCH, Longbrake EE, Popovich PG, Imai T, Combadiere C, Murphy PM, Wynick D: Intra-neural administration of fractalkine attenuates neuropathic pain-related behaviour. J Neurochem 2008,106(2):640–649.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Gadient RA, Otten UH: Interleukin-6 (IL-6) - A molecule with both beneficial and destructive potentials. Prog Neurobiol 1997,52(5):379–390.CrossRefPubMed Gadient RA, Otten UH: Interleukin-6 (IL-6) - A molecule with both beneficial and destructive potentials. Prog Neurobiol 1997,52(5):379–390.CrossRefPubMed
37.
Zurück zum Zitat Osamura N, Ikeda K, Ito T, Higashida H, Tomita K, Yokoyama S: Induction of interleukin-6 in dorsal root ganglion neurons after gradual elongation of rat sciatic nerve. Exp Neurol 2005,195(1):61–70.CrossRefPubMed Osamura N, Ikeda K, Ito T, Higashida H, Tomita K, Yokoyama S: Induction of interleukin-6 in dorsal root ganglion neurons after gradual elongation of rat sciatic nerve. Exp Neurol 2005,195(1):61–70.CrossRefPubMed
38.
Zurück zum Zitat Emson PC: In situ hybridization as a methodological tool for the neuroscientist. Trends Neurosci 1993,16(1):9–16.CrossRefPubMed Emson PC: In situ hybridization as a methodological tool for the neuroscientist. Trends Neurosci 1993,16(1):9–16.CrossRefPubMed
39.
Zurück zum Zitat Takeda M, Takahashi M, Matsumoto S: Contribution of the activation of satellite glia in sensory ganglia to pathological pain. Neurosci Biobehav Rev 2009,33(6):784–792.CrossRefPubMed Takeda M, Takahashi M, Matsumoto S: Contribution of the activation of satellite glia in sensory ganglia to pathological pain. Neurosci Biobehav Rev 2009,33(6):784–792.CrossRefPubMed
40.
Zurück zum Zitat Elson K, Simmons A, Speck P: Satellite cell proliferation in murine sensory ganglia in response to scarification of the skin. Glia 2004,45(1):105–109.CrossRefPubMed Elson K, Simmons A, Speck P: Satellite cell proliferation in murine sensory ganglia in response to scarification of the skin. Glia 2004,45(1):105–109.CrossRefPubMed
41.
Zurück zum Zitat Sommer C, Galbraith JA, Heckman HM, Myers RR: Pathology of experimental compression neuropathy producing hyperesthesia. J Neuropath Exp Neurol 1993,52(3):223–233.CrossRefPubMed Sommer C, Galbraith JA, Heckman HM, Myers RR: Pathology of experimental compression neuropathy producing hyperesthesia. J Neuropath Exp Neurol 1993,52(3):223–233.CrossRefPubMed
42.
Zurück zum Zitat Quintana A, Erta M, Ferrer B, Comes G, Giralt M, Hidalgo J: Astrocyte-specific deficiency of interleukin-6 and its receptor reveal specific roles in survival, body weight and behavior. Brain Behav Immun 2013,27(2):162–173.CrossRefPubMed Quintana A, Erta M, Ferrer B, Comes G, Giralt M, Hidalgo J: Astrocyte-specific deficiency of interleukin-6 and its receptor reveal specific roles in survival, body weight and behavior. Brain Behav Immun 2013,27(2):162–173.CrossRefPubMed
43.
Zurück zum Zitat Dubovy P, Klusakova I, Svizenska I, Brazda V: Satellite glial cells express IL-6 and corresponding signal-transducing receptors in the dorsal root ganglia of rat neuropathic pain model. Neuron Glia Biol 2010,6(1):73–83.CrossRefPubMed Dubovy P, Klusakova I, Svizenska I, Brazda V: Satellite glial cells express IL-6 and corresponding signal-transducing receptors in the dorsal root ganglia of rat neuropathic pain model. Neuron Glia Biol 2010,6(1):73–83.CrossRefPubMed
44.
Zurück zum Zitat Yu LC, Hansson P, BroddaJansen G, Theodorsson E, Lundeberg T: Intrathecal CGRP(8–37)-induced bilateral increase in hindpaw withdrawal latency in rats with unilateral inflammation. Brit J Pharmacol 1996,117(1):43–50.CrossRef Yu LC, Hansson P, BroddaJansen G, Theodorsson E, Lundeberg T: Intrathecal CGRP(8–37)-induced bilateral increase in hindpaw withdrawal latency in rats with unilateral inflammation. Brit J Pharmacol 1996,117(1):43–50.CrossRef
45.
Zurück zum Zitat Oaklander AL, Belzberg AJ: Unilateral nerve injury down-regulates mRNA for Na+ channel SCN10A bilaterally in rat dorsal root ganglia. Mol Brain Res 1997,52(1):162–165.CrossRefPubMed Oaklander AL, Belzberg AJ: Unilateral nerve injury down-regulates mRNA for Na+ channel SCN10A bilaterally in rat dorsal root ganglia. Mol Brain Res 1997,52(1):162–165.CrossRefPubMed
46.
Zurück zum Zitat Pitcher GM, Henry JL: Cellular mechanisms of hyperalgesia and spontaneous pain in a spinalized rat model of peripheral neuropathy: changes in myelinated afferent inputs implicated. Eur J Neurosci 2000,12(6):2006–2020.CrossRefPubMed Pitcher GM, Henry JL: Cellular mechanisms of hyperalgesia and spontaneous pain in a spinalized rat model of peripheral neuropathy: changes in myelinated afferent inputs implicated. Eur J Neurosci 2000,12(6):2006–2020.CrossRefPubMed
47.
Zurück zum Zitat Kleinschnitz C, Brinkhoff J, Zelenka M, Sommer C, Stoll G: The extent of cytokine induction in peripheral nerve lesions depends on the mode of injury and NMDA receptor signaling. J Neuroimmunol 2004,149(1–2):77–83.CrossRefPubMed Kleinschnitz C, Brinkhoff J, Zelenka M, Sommer C, Stoll G: The extent of cytokine induction in peripheral nerve lesions depends on the mode of injury and NMDA receptor signaling. J Neuroimmunol 2004,149(1–2):77–83.CrossRefPubMed
48.
Zurück zum Zitat Uceyler N, Tscharke A, Sommer C: Early cytokine expression in mouse sciatic nerve after chronic constriction nerve injury depends on calpain. Brain Behav Immun 2007,21(5):553–560.CrossRefPubMed Uceyler N, Tscharke A, Sommer C: Early cytokine expression in mouse sciatic nerve after chronic constriction nerve injury depends on calpain. Brain Behav Immun 2007,21(5):553–560.CrossRefPubMed
49.
Zurück zum Zitat Koltzenburg M, Wall PD, McMahon SB: Does the right side know what the left is doing? Trends Neurosci 1999,22(3):122–127.CrossRefPubMed Koltzenburg M, Wall PD, McMahon SB: Does the right side know what the left is doing? Trends Neurosci 1999,22(3):122–127.CrossRefPubMed
50.
Zurück zum Zitat Jancalek R, Dubovy P, Svizenska I, Klusakova I: Bilateral changes of TNF-alpha and IL-10 protein in the lumbar and cervical dorsal root ganglia following a unilateral chronic constriction injury of the sciatic nerve. J Neuroinflamm 2010.,7(11): doi:10.1186/1742-2094-7-11. Jancalek R, Dubovy P, Svizenska I, Klusakova I: Bilateral changes of TNF-alpha and IL-10 protein in the lumbar and cervical dorsal root ganglia following a unilateral chronic constriction injury of the sciatic nerve. J Neuroinflamm 2010.,7(11): doi:10.1186/1742-2094-7-11.
51.
Zurück zum Zitat Jancalek R, Svizenska I, Klusakova I, Dubovy P: Bilateral changes of IL-10 protein in lumbar and cervical dorsal root ganglia following proximal and distal chronic constriction injury of peripheral nerve. Neurosci Lett 2011,501(2):86–91.CrossRefPubMed Jancalek R, Svizenska I, Klusakova I, Dubovy P: Bilateral changes of IL-10 protein in lumbar and cervical dorsal root ganglia following proximal and distal chronic constriction injury of peripheral nerve. Neurosci Lett 2011,501(2):86–91.CrossRefPubMed
52.
Zurück zum Zitat Bennett GJ, Xie YK: A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988,33(1):87–107.CrossRefPubMed Bennett GJ, Xie YK: A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988,33(1):87–107.CrossRefPubMed
53.
Zurück zum Zitat Maves TJ, Pechman PS, Gebhart GF, Meller ST: Possible chemical contribution from chromic gut sutures produces disorders of pain sensation like those seen in man. Pain 1993,54(1):57–69.CrossRefPubMed Maves TJ, Pechman PS, Gebhart GF, Meller ST: Possible chemical contribution from chromic gut sutures produces disorders of pain sensation like those seen in man. Pain 1993,54(1):57–69.CrossRefPubMed
54.
Zurück zum Zitat Clatworthy AL, Illich PA, Castro GA, Walters ET: Role of peri-axonal inflammation in the development of thermal hyperalgesia and guarding behavior in a rat model of neuropathic pain. Neurosci Lett 1995,184(1):5–8.CrossRefPubMed Clatworthy AL, Illich PA, Castro GA, Walters ET: Role of peri-axonal inflammation in the development of thermal hyperalgesia and guarding behavior in a rat model of neuropathic pain. Neurosci Lett 1995,184(1):5–8.CrossRefPubMed
55.
Zurück zum Zitat Sotgiu ML, Biella G: Contribution of central sensitization to the pain-related abnormal activity in neuropathic rats. Somatosens Mot Res 2000,17(1):32–38.CrossRefPubMed Sotgiu ML, Biella G: Contribution of central sensitization to the pain-related abnormal activity in neuropathic rats. Somatosens Mot Res 2000,17(1):32–38.CrossRefPubMed
56.
Zurück zum Zitat Sotgiu ML, Biella G: Spinal neuron sensitization facilitates contralateral input in rats with peripheral mononeuropathy. Neurosci Lett 1998,241(2–3):127–130.CrossRefPubMed Sotgiu ML, Biella G: Spinal neuron sensitization facilitates contralateral input in rats with peripheral mononeuropathy. Neurosci Lett 1998,241(2–3):127–130.CrossRefPubMed
57.
Zurück zum Zitat Suzuki R, Rahman W, Rygh LJ, Webber M, Hunt SP, Dickenson AH: Spinal-supraspinal serotonergic circuits regulating neuropathic pain and its treatment with gabapentin. Pain 2005,117(3):292–303.CrossRefPubMed Suzuki R, Rahman W, Rygh LJ, Webber M, Hunt SP, Dickenson AH: Spinal-supraspinal serotonergic circuits regulating neuropathic pain and its treatment with gabapentin. Pain 2005,117(3):292–303.CrossRefPubMed
58.
Zurück zum Zitat English AW, Tigges J, Lennard PR: Anatomical organization of long ascending propriospinal neurons in the cat spinal cord. J Comp Neurol 1985,240(4):349–358.CrossRefPubMed English AW, Tigges J, Lennard PR: Anatomical organization of long ascending propriospinal neurons in the cat spinal cord. J Comp Neurol 1985,240(4):349–358.CrossRefPubMed
59.
Zurück zum Zitat Sallmann S, Juttler E, Prinz S, Petersen N, Knopf U, Weiser T, Schwaninger M: Induction of interleukin-6 by depolarization of neurons. J Neurosci 2000,20(23):8637–8642.PubMed Sallmann S, Juttler E, Prinz S, Petersen N, Knopf U, Weiser T, Schwaninger M: Induction of interleukin-6 by depolarization of neurons. J Neurosci 2000,20(23):8637–8642.PubMed
60.
Zurück zum Zitat Arvidson B: A study of the perineurial diffusion barrier of a peripheral ganglion. Acta Neuropathol 1979, 46:139–144.CrossRefPubMed Arvidson B: A study of the perineurial diffusion barrier of a peripheral ganglion. Acta Neuropathol 1979, 46:139–144.CrossRefPubMed
61.
Zurück zum Zitat Olsson Y: Studies on vascular permeability in peripheral nerves. 1. Distribution of circulating fluorescent serum albumin in normal, crushed and sectioned rat sciatic nerve. Acta Neuropathol 1966,7(1):1–15.CrossRefPubMed Olsson Y: Studies on vascular permeability in peripheral nerves. 1. Distribution of circulating fluorescent serum albumin in normal, crushed and sectioned rat sciatic nerve. Acta Neuropathol 1966,7(1):1–15.CrossRefPubMed
62.
Zurück zum Zitat Mellick R, Cavanagh JB: Extracellular space of normal peripheral nerve and normal skin as measured by radioactive sulfate in chicken. Exp Neurol 1967,18(2):224–227.CrossRefPubMed Mellick R, Cavanagh JB: Extracellular space of normal peripheral nerve and normal skin as measured by radioactive sulfate in chicken. Exp Neurol 1967,18(2):224–227.CrossRefPubMed
63.
Zurück zum Zitat St-Jacques B, Ma WY: Role of prostaglandin E2 in the synthesis of the pro-inflammatory cytokine interleukin-6 in primary sensory neurons: an in vivo and in vitro study. J Neurochem 2011,118(5):841–854.CrossRefPubMed St-Jacques B, Ma WY: Role of prostaglandin E2 in the synthesis of the pro-inflammatory cytokine interleukin-6 in primary sensory neurons: an in vivo and in vitro study. J Neurochem 2011,118(5):841–854.CrossRefPubMed
64.
Zurück zum Zitat Hide I, Tanaka M, Inoue A, Nakajima K, Kohsaka S, Inoue K, Nakata Y: Extracellular ATP triggers tumor necrosis factor-alpha release from rat microglia. J Neurochem 2000,75(3):965–972.CrossRefPubMed Hide I, Tanaka M, Inoue A, Nakajima K, Kohsaka S, Inoue K, Nakata Y: Extracellular ATP triggers tumor necrosis factor-alpha release from rat microglia. J Neurochem 2000,75(3):965–972.CrossRefPubMed
65.
Zurück zum Zitat Karanth S, Yang G, Yeh J, Richardson PM: Nature of signals that initiate the immune response during Wallerian degeneration of peripheral nerves. Exp Neurol 2006,202(1):161–166.CrossRefPubMed Karanth S, Yang G, Yeh J, Richardson PM: Nature of signals that initiate the immune response during Wallerian degeneration of peripheral nerves. Exp Neurol 2006,202(1):161–166.CrossRefPubMed
66.
Zurück zum Zitat Levin ME, Jin JG, Ji RR, Tong JF, Pomonis JD, Lavery DJ, Miller SW, Chiang LW: Complement activation in the peripheral nervous system following the spinal nerve ligation model of neuropathic pain. Pain 2008,137(1):182–201.CrossRefPubMed Levin ME, Jin JG, Ji RR, Tong JF, Pomonis JD, Lavery DJ, Miller SW, Chiang LW: Complement activation in the peripheral nervous system following the spinal nerve ligation model of neuropathic pain. Pain 2008,137(1):182–201.CrossRefPubMed
67.
Zurück zum Zitat Verderio C, Matteoli M: ATP in neuron-glia bidirectional signalling. Brain Res Rev 2011,66(1–2):106–114.CrossRefPubMed Verderio C, Matteoli M: ATP in neuron-glia bidirectional signalling. Brain Res Rev 2011,66(1–2):106–114.CrossRefPubMed
68.
Zurück zum Zitat Gu YP, Chen Y, Zhang XF, Li GW, Wang CY, Huang LYM: Neuronal soma-satellite glial cell interactions in sensory ganglia and the participation of purinergic receptors. Neuron Glia Biol 2010,6(1):53–62.CrossRefPubMedPubMedCentral Gu YP, Chen Y, Zhang XF, Li GW, Wang CY, Huang LYM: Neuronal soma-satellite glial cell interactions in sensory ganglia and the participation of purinergic receptors. Neuron Glia Biol 2010,6(1):53–62.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Chen Y, Zhang XF, Wang CY, Li GW, Gu YP, Huang LYM: Activation of P2X7 receptors in glial satellite cells reduces pain through downregulation of P2X3 receptors in nociceptive neurons. Proc Natl Acad Sci USA 2008,105(43):16773–16778.CrossRefPubMedPubMedCentral Chen Y, Zhang XF, Wang CY, Li GW, Gu YP, Huang LYM: Activation of P2X7 receptors in glial satellite cells reduces pain through downregulation of P2X3 receptors in nociceptive neurons. Proc Natl Acad Sci USA 2008,105(43):16773–16778.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Zhang X, Chen Y, Wang C, Huang LYM: Neuronal somatic ATP release triggers neuron-satellite glial cell communication in dorsal root ganglia. Proc Natl Acad Sci USA 2007,104(23):9864–9869.CrossRefPubMedPubMedCentral Zhang X, Chen Y, Wang C, Huang LYM: Neuronal somatic ATP release triggers neuron-satellite glial cell communication in dorsal root ganglia. Proc Natl Acad Sci USA 2007,104(23):9864–9869.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Kushnir R, Cherkas PS, Hanani M: Peripheral inflammation upregulates P2X receptor expression in satellite glial cells of mouse trigeminal ganglia: A calcium imaging study. Neuropharmacology 2011,61(4):739–746.CrossRefPubMed Kushnir R, Cherkas PS, Hanani M: Peripheral inflammation upregulates P2X receptor expression in satellite glial cells of mouse trigeminal ganglia: A calcium imaging study. Neuropharmacology 2011,61(4):739–746.CrossRefPubMed
72.
Zurück zum Zitat Fusaro M, Pelegrini-Da-Silva A, Araldi D, Parada CA, Tambeli CH: P2X3 and P2X2/3 receptors mediate mechanical hyperalgesia induced by bradykinin, but not by pro-inflammatory cytokines, PGE(2) or dopamine. Eur J Pharmacol 2010,649(1–3):177–182.CrossRef Fusaro M, Pelegrini-Da-Silva A, Araldi D, Parada CA, Tambeli CH: P2X3 and P2X2/3 receptors mediate mechanical hyperalgesia induced by bradykinin, but not by pro-inflammatory cytokines, PGE(2) or dopamine. Eur J Pharmacol 2010,649(1–3):177–182.CrossRef
73.
Zurück zum Zitat Chessell IP, Hatcher JP, Bountra C, Michel AD, Hughes JP, Green P, Egerton J, Murfin M, Richardson J, Peck WL: Disruption of the P2X(7) purinoceptor gene abolishes chronic inflammatory and neuropathic pain. Pain 2005,114(3):386–396.CrossRefPubMed Chessell IP, Hatcher JP, Bountra C, Michel AD, Hughes JP, Green P, Egerton J, Murfin M, Richardson J, Peck WL: Disruption of the P2X(7) purinoceptor gene abolishes chronic inflammatory and neuropathic pain. Pain 2005,114(3):386–396.CrossRefPubMed
74.
Zurück zum Zitat Shigemoto-Mogami Y, Koizumi S, Tsuda M, Ohsawa K, Kohsaka S, Inoue K: Mechanisms underlying extracellular ATP-evoked interleukin-6 release in mouse microglial cell line, MG-5. J Neurochem 2001,78(6):1339–1349.CrossRefPubMed Shigemoto-Mogami Y, Koizumi S, Tsuda M, Ohsawa K, Kohsaka S, Inoue K: Mechanisms underlying extracellular ATP-evoked interleukin-6 release in mouse microglial cell line, MG-5. J Neurochem 2001,78(6):1339–1349.CrossRefPubMed
75.
Zurück zum Zitat Vollmer-Conna U, Fazou C, Cameron B, Li H, Brennan C, Luck L, Davenport T, Wakefield D, Hickie I, Lloyd A: Production of pro-inflammatory cytokines correlates with the symptoms of acute sickness behaviour in humans. Psychol Med 2004,34(7):1289–1297.CrossRefPubMed Vollmer-Conna U, Fazou C, Cameron B, Li H, Brennan C, Luck L, Davenport T, Wakefield D, Hickie I, Lloyd A: Production of pro-inflammatory cytokines correlates with the symptoms of acute sickness behaviour in humans. Psychol Med 2004,34(7):1289–1297.CrossRefPubMed
76.
Zurück zum Zitat Watkins LR, Maier SF: Immune regulation of central nervous system functions: from sickness responses to pathological pain. J Intern Med 2005,257(2):139–155.CrossRefPubMed Watkins LR, Maier SF: Immune regulation of central nervous system functions: from sickness responses to pathological pain. J Intern Med 2005,257(2):139–155.CrossRefPubMed
77.
Zurück zum Zitat Czlonkowski A, Stein C, Herz A: Peripheral mechanisms of opioid antinociception in inflammation - involvement of cytokines. Eur J Pharmacol 1993,242(3):229–235.CrossRefPubMed Czlonkowski A, Stein C, Herz A: Peripheral mechanisms of opioid antinociception in inflammation - involvement of cytokines. Eur J Pharmacol 1993,242(3):229–235.CrossRefPubMed
78.
Zurück zum Zitat Wells MR, Racis SP, Vaidya U: Changes in plasma cytokines associated with peripheral nerve injury. J Neuroimmunol 1992,39(3):261–268.CrossRefPubMed Wells MR, Racis SP, Vaidya U: Changes in plasma cytokines associated with peripheral nerve injury. J Neuroimmunol 1992,39(3):261–268.CrossRefPubMed
79.
Zurück zum Zitat Brennan TJ, Vandermeulen EP, Gebhart GF: Characterization of a rat model of incisional pain. Pain 1996,64(3):493–501.CrossRefPubMed Brennan TJ, Vandermeulen EP, Gebhart GF: Characterization of a rat model of incisional pain. Pain 1996,64(3):493–501.CrossRefPubMed
80.
Zurück zum Zitat Seltzer Z, Dubner R, Shir Y: A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990,43(2):205–218.CrossRefPubMed Seltzer Z, Dubner R, Shir Y: A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990,43(2):205–218.CrossRefPubMed
81.
Zurück zum Zitat Takaishi K, Eisele JH, Carstens E: Behavioral and electrophysiological assessment of hyperalgesia and changes in dorsal horn responses following partial sciatic nerve ligation in rats. Pain 1996,66(2–3):297–306.CrossRefPubMed Takaishi K, Eisele JH, Carstens E: Behavioral and electrophysiological assessment of hyperalgesia and changes in dorsal horn responses following partial sciatic nerve ligation in rats. Pain 1996,66(2–3):297–306.CrossRefPubMed
82.
Zurück zum Zitat Jaggi AS, Singh N: Role of different brain areas in peripheral nerve injury-induced neuropathic pain. Brain Res 2011, 1381:187–201.CrossRefPubMed Jaggi AS, Singh N: Role of different brain areas in peripheral nerve injury-induced neuropathic pain. Brain Res 2011, 1381:187–201.CrossRefPubMed
83.
Zurück zum Zitat Saade NE, Jabbur SJ: Nociceptive behavior in animal models for peripheral neuropathy: Spinal and supraspinal mechanisms. Prog Neurobiol 2008,86(1):22–47.CrossRefPubMed Saade NE, Jabbur SJ: Nociceptive behavior in animal models for peripheral neuropathy: Spinal and supraspinal mechanisms. Prog Neurobiol 2008,86(1):22–47.CrossRefPubMed
84.
Zurück zum Zitat Pitcher GM, Ritchie J, Henry JL: Nerve constriction in the rat: model of neuropathic, surgical and central pain. Pain 1999,83(1):37–46.CrossRefPubMed Pitcher GM, Ritchie J, Henry JL: Nerve constriction in the rat: model of neuropathic, surgical and central pain. Pain 1999,83(1):37–46.CrossRefPubMed
85.
Zurück zum Zitat Zhong J, Dietzel ID, Wahle P, Kopf M, Heumann R: Sensory impairments and delayed regeneration of sensory axons in interleukin-6-deficient mice. J Neurosci 1999,19(11):4305–4313.PubMed Zhong J, Dietzel ID, Wahle P, Kopf M, Heumann R: Sensory impairments and delayed regeneration of sensory axons in interleukin-6-deficient mice. J Neurosci 1999,19(11):4305–4313.PubMed
86.
Zurück zum Zitat Bridges D, Thompson SWN, Rice ASC: Mechanism in neuropathic pain. Br J Anaesth 2001,87(1):12–26.CrossRefPubMed Bridges D, Thompson SWN, Rice ASC: Mechanism in neuropathic pain. Br J Anaesth 2001,87(1):12–26.CrossRefPubMed
87.
Zurück zum Zitat Wells MR, Vaidya U, Schwartz JP: Bilateral phasic increases in dorsal-root ganglia nerve growth-factor synthesis after unilateral sciatic-nerve crush. Exp Brain Res 1994,101(1):53–58.CrossRefPubMed Wells MR, Vaidya U, Schwartz JP: Bilateral phasic increases in dorsal-root ganglia nerve growth-factor synthesis after unilateral sciatic-nerve crush. Exp Brain Res 1994,101(1):53–58.CrossRefPubMed
88.
Zurück zum Zitat Yamaguchi H, Ochi M, Mori R, Ryoke K, Yamamoto S, Iwata A, Uchio Y: Unilateral sciatic nerve injury stimulates contralateral nerve regeneration. Neuroreport 1999,10(6):1359–1362.CrossRefPubMed Yamaguchi H, Ochi M, Mori R, Ryoke K, Yamamoto S, Iwata A, Uchio Y: Unilateral sciatic nerve injury stimulates contralateral nerve regeneration. Neuroreport 1999,10(6):1359–1362.CrossRefPubMed
89.
Zurück zum Zitat Ryoke K, Ochi M, Iwata A, Uchio Y, Yamamoto S, Yamaguchi H: A conditioning lesion promotes in vivo nerve regeneration in the contralateral sciatic nerve of rats. Biochem Biophys Res Commun 2000,267(3):715–718.CrossRefPubMed Ryoke K, Ochi M, Iwata A, Uchio Y, Yamamoto S, Yamaguchi H: A conditioning lesion promotes in vivo nerve regeneration in the contralateral sciatic nerve of rats. Biochem Biophys Res Commun 2000,267(3):715–718.CrossRefPubMed
Metadaten
Titel
Bilateral elevation of interleukin-6 protein and mRNA in both lumbar and cervical dorsal root ganglia following unilateral chronic compression injury of the sciatic nerve
verfasst von
Petr Dubový
Václav Brázda
Ilona Klusáková
Ivana Hradilová-Svíženská
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2013
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-10-55

Weitere Artikel der Ausgabe 1/2013

Journal of Neuroinflammation 1/2013 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Blutdrucksenkung schon im Rettungswagen bei akutem Schlaganfall?

31.05.2024 Apoplex Nachrichten

Der optimale Ansatz für die Blutdruckkontrolle bei Patientinnen und Patienten mit akutem Schlaganfall ist noch nicht gefunden. Ob sich eine frühzeitige Therapie der Hypertonie noch während des Transports in die Klinik lohnt, hat jetzt eine Studie aus China untersucht.

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Schutz der Synapsen bei Alzheimer

29.05.2024 Morbus Alzheimer Nachrichten

Mit einem Neurotrophin-Rezeptor-Modulator lässt sich möglicherweise eine bestehende Alzheimerdemenz etwas abschwächen: Erste Phase-2-Daten deuten auf einen verbesserten Synapsenschutz.

Hörschwäche erhöht Demenzrisiko unabhängig von Beta-Amyloid

29.05.2024 Hörstörungen Nachrichten

Hört jemand im Alter schlecht, nimmt das Hirn- und Hippocampusvolumen besonders schnell ab, was auch mit einem beschleunigten kognitiven Abbau einhergeht. Und diese Prozesse scheinen sich unabhängig von der Amyloidablagerung zu ereignen.