• 沒有找到結果。

Spinal p38 Activity and Analgesic Effect after Low- and High-intensity Electroacupuncture Stimulation in a Plantar Incision Rat Model

N/A
N/A
Protected

Academic year: 2021

Share "Spinal p38 Activity and Analgesic Effect after Low- and High-intensity Electroacupuncture Stimulation in a Plantar Incision Rat Model"

Copied!
1
0
0

加載中.... (立即查看全文)

全文

(1)

Category: Research Paper

Spinal p38 Activity and Analgesic Effect after Low and Highintensity Electroacupuncture Stimulation in a Plantar Incision Rat Model

Sheng-Feng Hsua,b,1, Yen-Jing Zenga,c,1, Shih-Ying Tsaid, Kuen-Bao Chene, Julia Yi-Ru Chenf,g, Ju-Hsin Changc,e, Yeong-Ray Wend,e,h

a Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical

University, Taichung, Taiwan

b Department of Acupuncture, China Medical University Hospital Taipei Branch, Taipei,

Taiwan

c Graduate Institute of Clinical Medical Science, College of Medicine, China Medical

University, Taichung, Taiwan

d Department of Anesthesiology, School of Medicine, China Medical University, Taichung,

Taiwan

e Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan f Department of Pediatrics, School of Medicine, Taipei Medical University, Taipei, Taiwan g Guang Li Biomedicine, Inc. ,Xizhi, New Taipei City, Taiwan

1 2

3 4

5

6 7 8

9

10 11

12 13

14 15

16 17 18 19

(2)

h Research Center for Chinese Medicine and Acupuncture, China Medical University, Taichung, Taiwan

1 S.-F. Hsu and Y.-J. Zeng have equal contributions as the first authors in this study.

Corresponding Author:

Yeong-Ray Wen, M.D., Ph.D.

Department of Anesthesiology, China Medical University Hospital

No. 2, Yuh-Der Rd, North District, 40447, Taichung, Taiwan

Tel.: +886-4-22052121 ext 3562;

Fax: +886-4-22052121 ext 3598.

*E-mail: yr.wen@yahoo.com.tw; yrwen@mail.cmu.edu.tw 20

21 22

23

24 25

26

27 28

29

30

31

(3)

Abstract

Aims: Postoperative pain is a major problem. Electroacupuncture (EA) has been accepted as a

useful and low-risk complementary therapy for post-operative pain. Animal studies indicate that surgical incision activates p38 MAPK in the spinal microglia, which critically contributes to post-incisional nociceptive development. How EA affects incision-induced p38 activation is important but yet to be fully elucidated.

Methods: Male adult rats received plantar incision (PI) at the right hind paw followed by 30-

min EA of 4-Hz, one of two intensities (3 and 10 mA), and at right ST36 (Zusanli) acupoint immediately after PI and for 3 successive days. EA analgesia was evaluated by von Frey fibers and Hargreaves’ tests. Spinal p38 activation was examined by immunostaining. In separate groups, SB203580, a p38 inhibitor, was intrathecally injected alone or with EA to test the combining effect on nociception and spinal phospho-p38.

Key findings: EA of 10-mA significantly ameliorated mechanical allodynia, but 3-mA did

not. None of them altered thermal hyperalgesia. Repeated EA could not inhibit phospho-p38 in the PI rats, contrarily, EA per se significantly induced phospho-p38 in the normal rats.

Intrathecal SB203580 injection dose-dependently prevented PI-induced allodynia.

Combination of low-dose SB203580 and 3-mA EA, which were ineffective individually, profoundly reduce post-PI allodynia.

32 33 34 35 36 37

38 39 40 41 42 43

44 45 46 47 48 49

(4)

Significance: We demonstrated that 10-mA EA exerts a significant inhibition against post-PI

mechanical hypersensitivity via a p38-independent pathway. Importantly, co-treatment with low-dose p38 inhibitor and 3-mA EA can counteract spinal phospho-p38 to exert strong analgesic effect. Our finding suggests a novel strategy to improve EA analgesic quality.

Keywords: electroacupuncture (EA); p38 MAPK; postoperative pain; Zusanli (ST36); spinal cord

50 51 52 53

54 55 56

57

(5)

Introduction

Post-operative pain is a common acute pain problem. Poorly controlled pain increases the risks of heart attack, pneumonia, deep vein thrombosis, immune impairment, anxiety, and persistent neuropathic pain (Kehlet et al. 2006; Liu and Wu 2007). Standard opioid administration remains the mainstay (Momeni et al. 2006), however, its use is complicated with opioid-related side effects which degrade analgesic quality. Our and other human studies demonstrated perioperative acupuncture or electroacupuncture (EA) could simultaneously reduce postoperative opioid consumption, spare adverse symptoms, and meanwhile maintain adequate analgesia (J. G. Lin et al. 2002; S. M. Wang et al. 2008b). Although EA has been accepted to pose these advantages, its use is still unpopular due to unclear mechanisms and low therapeutic efficacies.

Opioid-dependent and opioid-independent mechanisms interactively underlie acupuncture-induced analgesia (Han 2003; Sun et al. 2008; Shu Ming Wang et al. 2008a;

Wen et al. 2010). Following acupuncture stimulation, neuron-based releases of neuropeptides, including endogenous opiates-like endorphin, enkephalin, dynorphin, and endomorphin (Cabyoglu et al. 2006; Fukazawa et al. 2005; Han 2004, 2003; Han and Terenius 1982; Yun Wang et al. 2005); non-opioid substances like serotonin (Baek et al.

2005; Cabyoglu et al. 2006; Chang et al. 2004; J. G. Lin and Chen 2008), noradrenaline (Cabyoglu et al. 2006; Chang et al. 2004; Wen et al. 2010), oxytocin (Yang et al. 2007), 58

59 60 61 62 63 64 65 66 67 68

69 70 71 72 73 74 75 76

(6)

neuropeptide Y (Lee et al. 2009), neurotrophin-3 (Mi et al. 2011), somatostatin (Dong et al.

2005), spinal orexin A (Feng et al. 2012), and newly found peripheral release of adenosine (Goldman et al. 2010) were all involved to play roles in regulating acupuncture effects.

However, how these molecules participate and interact in various pain processes are complex and unclarified.

On the other hand, different lines of studies proved that neuroglia are powerful contributors or modulators in persistent pain states (Gosselin et al. 2010; Ji et al. 2009). Using a plantar incision (PI) model, which imitates postoperative pain as a preclinical animal study (Brennan et al. 1996), we showed that activation of p38 mitogen-activated protein kinase (MAPK) within microglia in the spinal dorsal horn contributed to nociceptive development at early post-PI stage (Wen et al. 2009). Pretreatment with p38 inhibitor suppressed phosphorylated-p38 (p-p38), an activated form of p38, can dose-dependently prevented behavioral hypersensitivity and downstream proinflammatory cytokines and chemokines (Lu et al. 2013; Wen et al. 2009). Emerging evidence indicated that EA effect may be partly mediated through microglial regulation by inhibiting intracellular signaling, including p-p38, and exocytosis of inflammatory mediators in many pain conditions (Gim et al. 2011; L. L.

Liang et al. 2010; Y. Liang et al. 2012; Mi et al. 2011; Shan et al. 2007; Sun et al. 2006; K.

D. Xu et al. 2010). Therefore, it would be of great value to investigate how EA controls p38 activation in surgical pain.

77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95

(7)

The present animal study aimed to answer the following questions: first, if EA could ameliorate PI-induced nociceptive behaviors; second, if EA suppresses incision-induced p- p38 in spinal microglia; and third, if combination of EA and p38 inhibitor could enhance analgesic effect.

96 97 98 99 100

(8)

Materials and Methods Experimental Animals

Adult male Sprague–Dawley rats (200–250 g; BioLASCO, Taiwan) were tri-housed and allowed to freely access food and water. The animal facility maintained a 12-h light-dark cycle at 22–24C and humidity of 70%. Animal managements were approved by Institutional Animal Care and Use Committee of China Medical University and followed the institutional Animal Care Guidelines. Efforts were made to minimize animal number and suffering.

Anesthesia, PI model, and EA

Experiments design and protocols were referring to our previous PI and EA studies (Brennan et al. 1996; Wen et al. 2009; Wen et al. 2007). In brief, the rats were anesthetized with 2%

isoflurane in oxygen via a nasal mask. An one-cm longitudinal incision was made at plantar surface of the right hind paw, 0.5 cm from the edge of the heel, through skin, plantar fascia, until flexor digitorum brevis muscle. The skin wound was sutured by skin layers with 50 nylon. Then the rats were transferred to transparent holders and were continuously anesthetized with 1% isoflurane in 100% oxygen via a cone nasal mask for EA stimulation.

EA was conducted at Zusanli (ST36) of right hind limb by a pair of 36G (0.22 mm in diameter) acupuncture needles with electric stimulation generated from a Grass S88 electrostimulator (Astromed, Grass, West Warwick, RI, USA) via constant current units 101

102

103 104 105 106 107

108 109

110 111 112 113 114 115 116

117 118 119

(9)

(Grass CCU1A, West Warwick, RI, USA). Stimulation parameters were set at 4-Hz square waves of 0.5 msec pulse width for a period of 30 minutes, following our lab protocol (Wen et al. 2007). Two intensities, 3 mA (low-intensity) and 10 mA (high-intensity), were applied to rats according to the group allocation. ST36 is the most frequently used acupoint and has been shown to produce systemic analgesic effects in animals (Pomeranz et al. 1977; Wen et al. 2007). The rats in the sham EA groups received needle insertion in muscle layer (5 mm depth) of ST36 but without electrostimulation. All rats were removed from the anesthetic apparatus after stop of EA, and all of them rapidly recovered to a movable state within 12 min. The recovery was so fast that anesthesia would not interfere the first behavioral test conducted 1 h later. We had shown this manipulation did not change behaviors or induce Fos expression (Wen et al. 2010).

Behavioral tests

Animals were acclimatized to experimental environment from at least 2 days prior to study.

To test mechanical threshold, the rats were put in inverted plastic boxes (10×10×20 cm) on an elevated mesh floor and allowed 30 min for habituation. Tactile thresholds were measured by von Frey fibers (Stoelting, Wood Dale, IL, USA). The paw was pressed with one of a series of von Frey fibers with logarithmically incrementing stiffness (0.4, 1.0, 2.0, 4.0, 6.0, 10.0, 15.0, and 26.0 g) perpendicularly onto the plantar medial surface. Each fiber was 120

121 122 123 124 125 126 127 128 129 130 131

132

133 134 135 136 137 138

(10)

applied for 5–6 sec. The 50% withdrawal threshold was determined using up-down method (Chaplan et al. 1994). The above protocol had been published (Gao et al. 2010; M. L. Lin et al. 2014). To test thermal thresholds, animals were put in a plastic box placed on a glass plate pre-warmed to a constant 30C (Plantar Test Apparatus, IITC, CA, USA), and the plantar surface was exposed to a beam of radiant heat underneath the glass floor. The baseline latencies were adjusted to 810 sec with a maximum of 20 sec as cut off to prevent potential heat injury. The latencies were averaged over three trials, separated by a 5-min interval (Hargreaves et al. 1988). The experimenter who performed these behavioral tests was blind to the group allocation of the rats.

Intrathecal (i.t.) administration

To evaluate the role of p-p38 in response to plantar incision and EA, the rats received intrathecal administration of a p38 inhibitor, SB203580 (Cell signaling Technology, Danvers, MA, USA). One hour before plantar incision, the rats were transiently anesthetized with high concentration of sevoflurane and back hairs were shaved to expose the lower back skin. After careful identification of L45 or L56 interspace, dural puncture to the intrathecal space was performed with a 30G needle and a Hamilton microsyringe (Hamilton Co., Nevada, USA).

Correct placement of needle was confirmed by a sign of brisk tail flick. Three doses of SB203580 (0.2, 0.5 or 2.0 mM in 10 μL) were slowly injected to the study groups. Because 139

140 141 142 143 144 145 146 147 148

149

150 151 152 153 154 155 156 157

(11)

SB203580 had to be dissolved in dimethyl sulfoxide (DMSO) which has anti-inflammatory (Gorog and Kovacs 1968) and analgesic properties (Evans et al. 1993), two concentrations of DMSO (2% and 20%) at the identical volume were injected as vehicle control to exclude bias of DMSO's potential effect.

Immunostaining

The animals were deeply anesthetized by isoflurane for transcardial perfusion of normal saline at room temperature (RT), followed by 4C, 4% paraformaldehyde in 0.1 M phosphate buffer (PB). The L45 spinal cord segments were carefully removed, post-fixed overnight, and cryoprotected in 30% sucrose/normal saline for 3 days. The transverse spinal sections were cut in cryostat (LEICA CM3050S, Nussloch, Germany) at a thickness of 30 μm and collected in 0.1 M PB. After blocking with 3% normal goat serum for 1 h at RT, the sections were incubated with primary antibodies containing 1% normal goat serum and 0.3% Triton X-100 overnight. Sections were then incubated with biotinylated anti-rabbit IgG (1:400, Vector Laboratories, Burlingame, CA, USA) and subsequently in an avidin-biotin-peroxidase complex/diaminiobenzidine-H2O2 solution (Elite ABC kit, Peroxidase substrate kit, Vector Laboratory). Spinal free-floating sections were mounted onto gelatin-coated glass slides, air- dried, dehydrated, cleared with xylene and coverslipped with Entellan mounting medium (Merck, Darmstadt, Germany). For double immunofluorescence, sections were incubated 158

159 160 161 162

163

164 165 166 167 168 169 170 171 172 173 174 175 176

(12)

with a mixture of two primary antibodies from different species at 4C, followed by a mixture of Cy3-conjugated (1:400, Jackson ImmunoResearch, West Grove, PA, USA) and Alexa Fluor 488-conjugated (1:400, Jackson ImmunoResearch) secondary antibodies at RT.

In this study, our primary antibodies include anti-phospho-p38 (rabbit, 1:400, Cell Signaling Technology), anti-neuronal nuclei (NeuN, mouse, 1:500, Millipore), anti-CD11B (OX-42, mouse, 1:50, Chemicon), and anti-glial fibrillary acidic protein (GFAP, mouse, 1:4000, Chemicon).

The stained sections were examined with Zeiss Axio Imager A2 microscopy (Göttingen, German) and images were captured with a CCD camera. At a magnification of x10, immunereactive (ir) cells in the dorsal horns were counted on randomly chosen sections, at least eight for each spinal segment from each rat, and then averaged for each rat data.

Experimental protocol

We first tested the behavioral changes in rats with plantar incision by inspecting their mechanical and heat thresholds over time (Fig. 1), then we tested effect of EA on PI-induced nociceptive hypersensitivities (Fig. 2). There were 4 groups: (1) the Naive: no treatment; (2) the PI+Sham: PI followed by repeated sham EA stimulations; (3) the PI+3 mA EA: PI with repeated 3-mA EA treatments; and (4) the PI+10 mA EA: PI with repeated 10-mA EA treatments. Behavioral tests were conducted at least 2 days before PI as baseline data and at 1 177

178 179 180 181 182 183 184 185 186 187

188

189 190 191 192 193 194 195

(13)

h, 3 h after PI on Day 0, and 1 h after EA on Day 1, 2, and 3. Spinal p-p38 expressions on the Day 3 were compared among groups and double-staining was conducted to identify p-p38-ir cell type (Fig. 3 and 4).

To understand if EA per se induces p-p38, the naïve rats received sham EA, 2-mA EA, or 10-mA EA (Fig. 5) for 4 days were sacrificed for immunostaining analysis. In separate groups, injections of p38 inhibitor at different doses were carried out to show effects of p38 activation on incisional pain (Fig. 6). One hour before PI, 10 L of (1) saline, (2) 20%

DMSO, (3) 0.2 mM SB203580 in 2% DMSO, (4) 0.5 mM SB203580 in 5% DMSO, or (5) 2.0 mM SB203580 in 20% DMSO were i.t. injected. Behavior responses were measured at 1 h, 3 h and one day after PI.

To test the combining effect of EA and p38 inhibitor, 5 groups were included (Fig. 7):

(1) Saline: a control group of pre-PI i.t saline injection and post-PI sham needling; (2) 2%

DMSO: a control group with pre-PI i.t. 2% DMSO and post-PI sham needling; (3) 0.2 mM SB: a group with pre-PI i.t. 0.2 mM SB203580 and post-PI sham needling; (4) 3 mA EA: a group with pre-PI i.t. saline and post-PI 3-mA EA; (5) SB+EA: a group with pre-PI 0.2 mM SB203580 and post-PI 3-mA EA. Behavioral tests were perfomed at post-PI 1 h, 3 h and on the next day.

Statistical Analysis 196

197 198 199 200 201 202 203 204 205

206 207 208 209 210 211 212

213 214

(14)

All data were presented as the mean ± S.E.M. Two-way analysis of variance (ANOVA) was conducted to analyze the influence among factors of time and treatments. Repeated- measurement ANOVA (RM-ANOVA) was conducted in the control group to assess effect of time course. One-way ANOVA with post hoc Tukey’s test was conducted to compare differences among groups. Software of PASW Statistics for Windows, version 18.0 (SPSS Inc, Chicago) was used for analysis. Statistical significance is considered when p < 0.05.

215 216 217 218 219 220

(15)

Results

Nociceptive hypersensitivity after plantar incision

PI resulted in mechanical allodynia and heat hyperalgesia immediately after surgery and persisted for about one week (Fig. 1), a similar finding to previous studies (Ririe et al. 2003;

Wen et al. 2009). Fifty-percent mechanical thresholds significantly decreased from pre-PI baseline (BL) 23.77  0.54 g to 0.49  0.05 g at 1 h and 0.65  0.09 g at 3 h (p < 0.001 vs.

BL), remained low till Day 3 (3.61  0.41 g, p < 0.001 vs. BL), and gradually increased from D5 to D7 (13.56  0.61 g, p < 0.001 vs. BL, Fig. 1A).

Thermal hypersensitivity showed similar changes (Fig. 1B). Mean withdrawal latency before incision was 9.50  0.61 s. Significant decrease occurred at post-PI 1 h (2.02  0.24 s, p < 0.001 vs. BL) through Day 3 (4.78  0.53 s, p < 0.001) to near baseline on Day 5 (7.10  0.23 s, p = 0.101). The two tests proved incision leads to an acute, strong painful sensation which recovered rapidly within few days, as clinical patients. Therefore, we limited observation within 3 days in subsequent experiments.

High-intensity EA suppresses PI-induced mechanical hypersensitivity

EA could reduce incisional pain on the paw depending on EA stimulating intensities (Fig.

2A). High-intensity EA (10 mA) significantly reduced mechanical allodynia since the first hour post PI, and kept significantly higher thresholds than those in the sham (PI+Sham) and 221

222

223 224 225 226 227 228

229 230 231 232 233 234

235 236

237 238 239

(16)

low-intensity EA (PI+3 mA EA) groups thereafter. Low-intensity EA (PI+3 mA EA group) did not produce any anti-allodynic effect throughout the course (p > 0.05 vs. PI+Sham).

Notably, neither 3-mA nor 10-mA EA could alter PI-induced heat hyperalgesia at any time point (Fig. 2B). Accordingly, higher strength EA is necessary to attenuate PI-induced mechanical allodynia and repeated treatment is required to maintain the effect. However, EA has no analgesic effect on heat hyperalgesia.

Repeated EA does not reduce PI-induced spinal p-p38

We found that PI drastically increased the amount of p-p38-ir cells in the spinal dorsal horn at 3 days after surgery (Fig. 3A), which spreaded diffusely without lamina differences.

However, to our astonishment, EA treatment did not decrease p-p38 expression in the dorsal horn (Fig. 3B and 3C). Quantitative analysis revealed that the 3-mA EA group did not differ from the PI+sham group, whereas the 10-mA EA group showed mildly higher p-p38-ir cell counts than those in the sham group (84.71  4.52 vs. 74.60  3.57, respectively; p = 0.049;

Fig. 3D).

p-p38 is localized within spinal microglia

We used immunofluorescent double staining to clearly identify the cells type of PI-induced p- p38. As shown in Fig. 4, a majority of p-p38 (red spots at panels A, B, C) were colocalized 240

241 242 243 244 245 246

247

248 249 250 251 252 253 254 255

256

257 258

(17)

with OX-42-positive microglia (Fig 4B’, 4B’’). No coexpression of p-p38 with NeuN- positive neuron (Fig. 4A’, A’’) or GFAP-positive astrocyte (Fig. 4C’, C’’) could be found.

This finding is correlated with previous studies in incisional and neuropathic pain model (Ito et al. 2009; Jin et al. 2003; Wen et al. 2009).

EA per se activates spinal p38 phosphorylation

Whether EA per se induced spinal p-p38 was also examined by repeated EA stimulations in the normal rats. We found that the naïve rats (without any noxious stimulation) showed very few p-p38-ir cells in the dorsal horn (about 20/section, Fig. 5A), whereas the EA-treated rats, though without vigorous PI injury, had clear p-p38-ir cells in dorsal horns (Fig. 5B and C).

Many EA-induced p-p38-ir cells are clustered at the medial half of the superficial laminae.

All p-p38-ir cells in Fig. 5A-C, no matter in naive or 2 EA groups, show small cell bodies and small, thin, and ramified processes, a state close to a “resting” condition in comparison with those with “activated” images in Fig. 3AC. Quantitative analysis revealed that either the 3-mA or 10-mA groups could significantly increase p-p38-ir cells more than that in the naive group (both p < 0.001; Fig. 5D).

p38 inhibitor dose-dependently suppresses mechanical allodynia 259

260 261 262 263

264

265 266 267 268 269 270 271 272 273 274 275

276

(18)

As predicted, p38 inhibitor effectively prevented nociceptive hypersensitivity. SB203580, when given at 1 h before PI, led to a dose-dependent suppression on mechanical allodynia for at least 3 h (3.33  0.65, 9.12  0.00, and 10.27  0.55 respectively for 0.2 mM, 0.5 mM, and 2.0 mM vs. 1.94  0.47 for saline, Fig. 6A), but all effects disappeared on the next day.

Particularly, SB203580 did not suppress heat hyperalgesia (all p > 0.05; Fig. 6B), indicating p-p38-mediated pathway may be irrelevant to heat nociception. The vehicle solvent with the highest concentration, 20% DMSO, did not have notable effect on mechanical pain but mildly inhibited heat pain (20% DMSO vs. Saline at 1 h; p = 0.014).

Low-dose p38 Inhibitor enhances low-intensity EA analgesia

Since EA per se was found to induce spinal p-p38 expression, it was deduced that inhibiting p38 activation might enhance EA analgesia. We knew that either 3-mA EA or 0.2 mM SB203580 did not produce obvious analgesia (Fig. 2A and 6A). Therefore, we hypothesized that combination of low-intensity EA and low-dose SB203580 may elicit a synergistic effect.

As expectedly, a significant enhancement of anti-allodynic effect was observed at 1 h and 3 h of Day 0 and Day 1, compared to the saline, 2% DMSO, and treated groups (all p < 0.001, Fig. 7A). Such combination showed an enhancement and prolongation of analgesia, with values greater than that of 10-mA EA (Fig. 2A) and that of 2.0 mM SB203580 (Fig. 6A). Of 277

278 279 280 281 282 283 284 285

286

287 288 289 290 291 292 293 294

(19)

note, the combination did not exert any additive effect on heat hyperalgesia (p > 0.05, Fig.

7B).

295 296

(20)

Discussion

In the present study, we demonstrated that 4-Hz EA at a high-intensity significantly reduces post-operative mechanical allodynia and repeated applications maintain analgesic effect. We also found EA at a low-intensity has no effect on incision-induced pain. More importantly, EA per se, either at a low or high intensity, activates p38 in spinal microglia in normal rats.

Co-treatment of low-intensity EA and low-dose p38 inhibitor could remarkably prolongs and enhances effect of low-intensity EA possibly via inhibiting EA-induced p38 activation.

Growing evidence proved that spinal p38 activation plays a pivotal role in pain initiation and development in neuropathic, inflammatory, or surgical pain (Ji et al. 2009; Ji and Suter 2007; Svensson et al. 2003; Wen et al. 2007; Zhuang et al. 2007). Spinal p38 activation is mainly expressed within microglia and is usually associated with microgliosis (Hanisch and Kettenmann 2007; Ji and Suter 2007). These changes start early after peripheral injuries and temporally parallel with progression of early nociceptive behaviors. Activated spinal microglia release pro-inflammatory cytokines and chemokines, further contribute to nociceptive hypersensitivities. Therefore, blockade of microglia-p38-cytokine-mediated signal pathways with agents such as p38 inhibitors, minocycline, or TNF inhibitors could reduce nociceptive processes (Ji and Suter 2007; Palladino et al. 2003; Zhuang et al. 2007).

There were studies demonstrating that EA could downregulate p-p38 or inhibit microglia in monoarthritic pain (Shan et al. 2007; Sun et al. 2008) and complete Freund's adjuvant (CFA)- 297

298 299 300 301 302 303

304 305 306 307 308 309 310 311 312 313 314 315

(21)

induced pain (Hargreaves et al. 1988; Mi et al. 2011; K. D. Xu et al. 2010). However, whether these p-p38 reductions were due to a direct action of EA or secondary to inhibition of pre- or post-synaptic noci-responsive neurons has not yet been clarified.

A striking finding of this study is that EA per se stimulates p38 activation in the ipsilateral dorsal horns, which is in contradiction with our original hypothesis and is opposed to published studies regarding to EA effects on MAPK activations. For instance, repeated daily EA at alternating-frequencies (2 and 100 Hz) could decrease CFA-induced paw swelling, mechanical hypersensitivity, and spinal p-p38 at 3 and 14 days after CFA injection (Y. Liang et al. 2012). The authors advocated that EA owned an inhibitory effect on p-p38- mediated inflammation and hyperalgesia. Another study demonstrated that repeated low- frequent EA produced synergistic inhibition against CFA-induced heat hyperalgesia for 2 days when co-injected with dizocilpine, a NMDA antagonist (Jang et al. 2011). As EA or dizocilpine was given alone, only EA inhibited CFA-induced p38 activation. They therefore concluded that EA-mediated p-p38 inhibition is NMDA-independent. Both studies similarly indicated the importance of p-p38 in inflammatory pain and effect of EA on spinal p38 activation. Nevertheless, none of them had examined EA-evoked p-p38 activation in normal rats or co-administered p38 inhibitor for verification.

Paw incision shares many molecular reactions in common with those after CFA injection, however, discrepancies are present between them. Incision seems to result in immediate, 316

317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332

333 334

(22)

excruciating pain much stronger than CFA-induced inflammatory pain. In this study, 3-mA EA is insufficient to reduce incision-induced allodynia, whereas low-intense EA (less than 2 mA) produced analgesia in many CFA studies (Huang et al. 2004; Jang et al. 2011; Lao et al.

2004; Y. Liang et al. 2012; R. X. Zhang et al. 2005b; Y. Zhang et al. 2011). In tail-flick and formalin-induced pain models, we demonstrated stronger EA stimulation (i.e. 20 times of muscle twitch intensity or above) led to stronger suppression, but meanwhile higher Fos expression in the spinal dorsal horns (Wen et al. 2010; Wen et al. 2007). Intense electrical stimulus sufficient to activate A and C fibers or at a pain-induction level, could also result in systemic/heterotopic analgesia in animals (Danziger et al. 2001; Morton et al. 1988; Romita and Henry 1996; Romita et al. 1997) and in humans (Roby-Brami et al. 1987; W. D. Xu et al.

2003). Collectively, strong EA stimulation may be required to suppress PI-induced strong pain, but may simultaneously trigger unwanted activations of MAPK system.

Of particular notice, we present that both 3-mA EA and 10-mA EA stimulation for 4 days activate p-p38 within spinal microglia in the normal rats, which has yet to be reported before.

Comparing to reports showing that EA below 2 mA suppressed p-p38 in CFA-induced pain, it is hypothesized that 3-mA EA strength is a boundary to determine the activation of microglial p38. When EA stimulation at strength above this level, A and C afferents may be activated to sensitize spinal noci-responsive neurons and MAPK phosphorylation. Besides, EA has been found to regulate immune response via recruitment of MAPK (p-ERK and p- 335

336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353

(23)

p38) activations in spleenic T helper cells (K. Wang et al. 2009). On the other hand, induction of endogenous opioids and binding to -, -, and maybe -opioid receptors following EA treatment is a well-proved acupuncture analgesic mechanism (Han 2003; R. X. Zhang et al.

2004). Of many inflammatory pain conditions, low-frequent EA analgesia could be completely reversed by - or -opioid receptor antagonists or toxin for -opioid receptor (Huang et al. 2004; R. X. Zhang et al. 2004; R. X. Zhang et al. 2005a; Y. Zhang et al. 2011).

In PI model, EA analgesia was completely reversed by systemic naloxone injection (Oliveira and Prado 2000) and daily EA effect was abolished by repeated naloxone injections in our recent study (Zeng et al. 2014). Because EA-induced analgesia is intensity-dependent (Wen et al. 2010), it is thus rational to assume that "primary" opioid-dependent EA analgesia could be counteracted by concomitant recruitment of p-p38-dependent nociceptive cascades as stimulation intensity is increased. Taken together, phenotype of EA analgesia under this circumstance could be a balance between opioid-mediated anti-nociception and p-p38- pertained drawback.

The results of co-administration of EA and p38 inhibitor justify our above deduction.

When given alone, 3-mA EA and 0.2-mM SB203580 were ineffective individually. The co- administration not only magnified 3-mA EA from ineffective to strong analgesia, but also prolonged the effect to the next day. Our finding is in consistence with the concept that silencing microglial p38 activation can reduce early neuroplastic sensitization and prevent 354

355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372

(24)

nociceptive development (Ji et al. 2013; Wen et al. 2011). Pretreatment with sufficient doses of SB203586 or FR167653, two different p38 inhibitors, produced 2-3 days of anti-allodynic effect on incision pain (Mizukoshi et al. 2013; Wen et al. 2009) and inflammatory pain (Li et al. 2010). Taken together, we believe that co-treatment could efficaciously overcome disadvantage of low analgesic potency in EA.

Here, we noted that EA did not inhibit heat hyperalgesia. Such differential analgesia for different pain patterns is possible. The anti-allodynic effect of this study is in agreement with previous PI study (Oliveira and Prado 2000) and consistent with a CFA study showing attenuation of mechanical, but not thermal hyperalgesia (Huang et al. 2004). However, there were other studies advocated that EA reduced heat hyperalgesia in CFA model (Jang et al.

2011; R. X. Zhang et al. 2005b). It is difficult to compare different results based on distinct study designs and pain models because EA actions include either inhibition or dis-inhibition of cellular signals and transmission pathways, and exert distinct functions among acute or persistent inflammatory pain (Huang et al. 2004; Lao et al. 2004; Y. Liang et al. 2012; R. X.

Zhang et al. 2005b), cancer pain (Kawasaki et al. 2008), ankle sprain pain (Koo et al. 2002), monoarthritis pain (Sun et al. 2008; Sun et al. 2006), and neuropathic pain (Dai et al. 2001;

Kim et al. 2005; Lau et al. 2008). In-depth exploration of activated mechanisms and standarization of EA protocols are required before comparing the analgesic variations.

373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391

(25)

Conclusions

In conclusion, an inference is drawn from the present study that strong EA stimulation is necessary to produce strong analgesic effect but is possibly associated with p38 activation in spinal microglia. EA analgesia may be depressed by concomitant p38 activation. We suggested that instead of using strong EA stimulation, co-administration of low-intensity EA with p38 inhibitor may provide better chances to potentiate EA efficacy and improve pain control quality.

Conflicts of Interest

All authors declared that there is no conflict of interest in this study. Part of our results had been reported as an oral presentation form in the annual meeting of the 16th International Congress of Oriental Medicine (ICOM).

392 393 394 395 396 397 398

399

400 401 402 403

404

(26)

Acknowledgments

We have especial thanks to Prof. Yi-Wen Lin (Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan) for his critical reviewing of this manuscript.

We also thank Miss Ya-Hsin Lou for her technical and official assistance. This study was sponsored by research grants from the National Science Council in Taiwan (NSC97-2314-B- 341-002-MY3, NSC101-2314-B-039-005-MY3), in part from China Medical University under the Aim for Top University Plan of the Ministry of Education in Taiwan to Y.-R. Wen, and the Joint Research Grant of Shin-Kong Wu Ho-Su Memorial Hospital and Taipei Medical University (SKH-TMU-98-15) to Y.-R. Wen and Julia Y.-R. Chen.

405 406 407 408 409 410 411 412 413

414

(27)

References

Baek YH, Choi DY, Yang HI, Park DS. Analgesic effect of electroacupuncture on inflammatory pain in the rat model of collagen-induced arthritis: mediation by cholinergic and serotonergic receptors. Brain Res 2005; 1057; 181-5.

Brennan TJ, Vandermeulen EP, Gebhart GF. Characterization of a rat model of incisional pain. Pain 1996; 64; 493-501.

Cabyoglu MT, Ergene N, Tan U. The mechanism of acupuncture and clinical applications.

Int J Neurosci 2006; 116; 115-25.

Chang FC, Tsai HY, Yu MC, Yi PL, Lin JG. The central serotonergic system mediates the analgesic effect of electroacupuncture on ZUSANLI (ST36) acupoints. J Biomed Sci 2004; 11; 179-85.

Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Meth 1994; 53; 55-63.

Dai Y, Kondo E, Fukuoka T, Tokunaga A, Miki K, Noguchi K. The effect of electroacupuncture on pain behaviors and noxious stimulus-evoked Fos expression in a rat model of neuropathic pain. J Pain 2001; 2; 151-9.

Danziger N, Gautron M, Le Bars D, Bouhassira D. Activation of diffuse noxious inhibitory controls (DNIC) in rats with an experimental peripheral mononeuropathy. Pain 2001;

91; 287-96.

Dong ZQ, Xie H, Ma F, Li WM, Wang YQ, Wu GC. Effects of electroacupuncture on expression of somatostatin and preprosomatostatin mRNA in dorsal root ganglions and spinal dorsal horn in neuropathic pain rats. Neuroscience letters 2005; 385; 189- 94.

Evans MS, Reid KH, Sharp JB, Jr. Dimethylsulfoxide (DMSO) blocks conduction in peripheral nerve C fibers: a possible mechanism of analgesia. Neurosci Lett 1993;

150; 145-8.

Feng XM, Mi WL, Xia F, Mao-Ying QL, Jiang JW, Xiao S, Wang ZF, Wang YQ, Wu GC.

Involvement of spinal orexin A in the electroacupuncture analgesia in a rat model of post-laparotomy pain. BMC Complement Altern Med 2012; 12; 225.

Fukazawa Y, Maeda T, Hamabe W, Kumamoto K, Gao Y, Yamamoto C, Ozaki M, Kishioka S. Activation of spinal anti-analgesic system following electroacupuncture stimulation in rats. J Pharmacol Sci 2005; 99; 408-14.

Gao YJ, Xu ZZ, Liu YC, Wen YR, Decosterd I, Ji RR. The c-Jun N-terminal kinase 1 (JNK1) in spinal astrocytes is required for the maintenance of bilateral mechanical allodynia under a persistent inflammatory pain condition. Pain 2010; 148; 309-19.

415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449

(28)

Gim GT, Lee JH, Park E, Sung YH, Kim CJ, Hwang WW, Chu JP, Min BI.

Electroacupuncture attenuates mechanical and warm allodynia through suppression of spinal glial activation in a rat model of neuropathic pain. Brain Res Bull 2011; 86;

403-11.

Goldman N, Chen M, Fujita T, Xu Q, Peng W, Liu W, Jensen TK, Pei Y, Wang F, Han X, Chen JF, Schnermann J, Takano T, Bekar L, Tieu K, Nedergaard M. Adenosine A1 receptors mediate local anti-nociceptive effects of acupuncture. Nat Neurosci 2010;

13; 883-8.

Gorog P, Kovacs IB. Effect of dimethyl sulfoxide (DMSO) on various experimental inflammations. Curr Ther Res Clin Exp 1968; 10; 486-92.

Gosselin RD, Suter MR, Ji RR, Decosterd I. Glial cells and chronic pain. Neuroscientist 2010; 16; 519-31.

Han JS. Acupuncture and endorphins. Neuroscience letters 2004; 361; 258-61.

Han JS. Acupuncture: neuropeptide release produced by electrical stimulation of different frequencies. Trends in Neurosciences 2003; 26; 17-22.

Han JS, Terenius L. Neurochemical basis of acupuncture analgesia. Annu Rev Pharmacol Toxicol 1982; 22; 193-220.

Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci 2007; 10; 1387-94.

Hargreaves K, Dubner R, Brown F, Flores C, Joris J. A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 1988; 32; 77-88.

Huang C, Hu Z-P, Long H, Shi Y-S, Han J-S, Wan Y. Attenuation of mechanical but not thermal hyperalgesia by electroacupuncture with the involvement of opioids in rat model of chronic inflammatory pain. Brain Research Bulletin 2004; 63; 99-103.

Ito N, Obata H, Saito S. Spinal microglial expression and mechanical hypersensitivity in a postoperative pain model: comparison with a neuropathic pain model. Anesthesiology 2009; 111; 640-8.

Jang JY, Kim HN, Koo ST, Shin HK, Choe ES, Choi BT. Synergistic antinociceptive effects of N-methyl-D-aspartate receptor antagonist and electroacupuncture in the complete Freund's adjuvant-induced pain model. Int J Mol Med 2011; 28; 669-75.

Ji RR, Berta T, Nedergaard M. Glia and pain: is chronic pain a gliopathy? Pain 2013; 154 Suppl 1; S10-28.

Ji RR, Gereau RWt, Malcangio M, Strichartz GR. MAP kinase and pain. Brain Res Rev 2009; 60; 135-48.

Ji RR, Suter MR. p38 MAPK, microglial signaling, and neuropathic pain. Mol Pain 2007; 3;

33.

450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485

(29)

Jin SX, Zhuang ZY, Woolf CJ, Ji RR. p38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J Neurosci 2003; 23; 4017-22.

Kawasaki Y, Zhang L, Cheng JK, Ji RR. Cytokine mechanisms of central sensitization:

distinct and overlapping role of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in regulating synaptic and neuronal activity in the superficial spinal cord.

J Neurosci 2008; 28; 5189-94.

Kehlet H, Jensen TS, Woolf CJ. Persistent postsurgical pain: risk factors and prevention.

Lancet 2006; 367; 1618-25.

Kim SK, Park JH, Bae SJ, Kim JH, Hwang BG, Min BI, Park DS, Na HS. Effects of electroacupuncture on cold allodynia in a rat model of neuropathic pain: mediation by spinal adrenergic and serotonergic receptors. Exp Neurol 2005; 195; 430-6.

Koo ST, Park YI, Lim KS, Chung K, Chung JM. Acupuncture analgesia in a new rat model of ankle sprain pain. Pain 2002; 99; 423-31.

Lao L, Zhang RX, Zhang G, Wang X, Berman BM, Ren K. A parametric study of electroacupuncture on persistent hyperalgesia and Fos protein expression in rats.

Brain Research 2004; 1020; 18-29.

Lau WK, Chan WK, Zhang JL, Yung KK, Zhang HQ. Electroacupuncture inhibits cyclooxygenase-2 up-regulation in rat spinal cord after spinal nerve ligation.

Neuroscience 2008.

Lee B, Shim I, Lee HJ, Yang Y, Hahm DH. Effects of acupuncture on chronic corticosterone- induced depression-like behavior and expression of neuropeptide Y in the rats.

Neuroscience letters 2009; 453; 151-6.

Li K, Lin T, Cao Y, Light AR, Fu KY. Peripheral Formalin Injury Induces 2 Stages Of Microglial Activation in the Spinal Cord. J Pain 2010.

Liang LL, Yang JL, Lu N, Gu XY, Zhang YQ, Zhao ZQ. Synergetic analgesia of propentofylline and electroacupuncture by interrupting spinal glial function in rats.

Neurochem Res 2010; 35; 1780-6.

Liang Y, Fang JQ, Du JY, Fang JF. Effect of Electroacupuncture on Activation of p38MAPK in Spinal Dorsal Horn in Rats with Complete Freund's Adjuvant-Induced Inflammatory Pain. Evid-Based Compl Alt 2012; 2012; 568273.

Lin JG, Chen WL. Acupuncture analgesia: a review of its mechanisms of actions. Am J Chin Med 2008; 36; 635-45.

Lin JG, Lo MW, Wen YR, Hsieh CL, Tsai SK, Sun WZ, Lin JG, Lo MW, Wen YR, Hsieh CL, Tsai SK, Sun WZ. The effect of high and low frequency electroacupuncture in pain after lower abdominal surgery. Pain 2002; 99; 509-14.

Lin ML, Lin WT, Huang RY, Chen TC, Huang SH, Chang CH, Tsai SY, Chiu HW, Yeh GC, Lin CW, Wen Y, R. Pulse radiofrequency inhibited activation of spinal mitogen- 486

487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523

(30)

activated protein kinases and ameliorated early neuropathic pain in rats. Eur J Pain 2014; 18; 659-70.

Liu SS, Wu CL. Effect of postoperative analgesia on major postoperative complications: a systematic update of the evidence. Anesth Analg 2007; 104; 689-702.

Lu Y, Zhao LX, Cao DL, Gao YJ. Spinal injection of docosahexaenoic acid attenuates carrageenan-induced inflammatory pain through inhibition of microglia-mediated neuroinflammation in the spinal cord. Neuroscience 2013; 241; 22-31.

Mi WL, Mao-Ying QL, Wang XW, Li X, Yang CJ, Jiang JW, Yu J, Wang J, Liu Q, Wang YQ, Wu GC. Involvement of spinal neurotrophin-3 in electroacupuncture analgesia and inhibition of spinal glial activation in rat model of monoarthritis. J Pain 2011; 12;

974-84.

Mizukoshi K, Sasaki M, Izumi Y, Miura M, Watanabe M, Amaya F. Activation of p38 mitogen-activated protein kinase in the dorsal root ganglion contributes to pain hypersensitivity after plantar incision. Neuroscience 2013; 234C; 77-87.

Momeni M, Crucitti M, De Kock M. Patient-controlled analgesia in the management of postoperative pain. Drugs 2006; 66; 2321-37.

Morton CR, Du HJ, Xiao HM, Maisch B, Zimmermann M. Inhibition of nociceptive responses of lumbar dorsal horn neurones by remote noxious afferent stimulation in the cat. Pain 1988; 34; 75-83.

Oliveira R, Prado WA. Anti-hyperalgesic effect of electroacupuncture in a model of post- incisional pain in rats. Braz J Med Biol Res 2000; 33; 957-60.

Palladino MA, Bahjat FR, Theodorakis EA, Moldawer LL. Anti-TNF-alpha therapies: the next generation. Nat Rev Drug Discov 2003; 2; 736-46.

Pomeranz B, Cheng R, Law P. Acupuncture reduces electrophysiological and behavioral responses to noxious stimuli: pituitary is implicated. Exp Neurol 1977; 54; 172-8.

Ririe DG, Vernon TL, Tobin JR, Eisenach JC, Ririe DG, Vernon TL, Tobin JR, Eisenach JC.

Age-dependent responses to thermal hyperalgesia and mechanical allodynia in a rat model of acute postoperative pain. Anesthesiology 2003; 99; 443-8.

Roby-Brami A, Bussel B, Willer JC, Le Bars D. An electrophysiological investigation into the pain-relieving effects of heterotopic nociceptive stimuli. Probable involvement of a supraspinal loop. Brain 1987; 110 ( Pt 6); 1497-508.

Romita VV, Henry JL. Intense peripheral electrical stimulation differentially inhibits tail vs.

limb withdrawal reflexes in the rat. Brain Res 1996; 720; 45-53.

Romita VV, Suk A, Henry JL. Parametric studies on electroacupuncture-like stimulation in a rat model: effects of intensity, frequency, and duration of stimulation on evoked antinociception. Brain Res Bull 1997; 42; 289-96.

Shan S, Qi-Liang MY, Hong C, Tingting L, Mei H, Haili P, Yan-Qing W, Zhi-Qi Z, Yu-Qiu Z. Is functional state of spinal microglia involved in the anti-allodynic and anti- 524

525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561

(31)

hyperalgesic effects of electroacupuncture in rat model of monoarthritis? Neurobiol Dis 2007; 26; 558-68.

Sun S, Cao H, Han M, Li TT, Zhao ZQ, Zhang YQ. Evidence for suppression of electroacupuncture on spinal glial activation and behavioral hypersensitivity in a rat model of monoarthritis. Brain Res Bull 2008; 75; 83-93.

Sun S, Chen WL, Wang PF, Zhao ZQ, Zhang YQ. Disruption of glial function enhances electroacupuncture analgesia in arthritic rats. Exp Neurol 2006; 198; 294-302.

Svensson CI, Marsala M, Westerlund A, Calcutt NA, Campana WM, Freshwater JD, Catalano R, Feng Y, Protter AA, Scott B, Yaksh TL. Activation of p38 mitogen- activated protein kinase in spinal microglia is a critical link in inflammation-induced spinal pain processing. J Neurochem 2003; 86; 1534-44.

Wang K, Wu H, Wang G, Li M, Zhang Z, Gu G. The effects of electroacupuncture on TH1/TH2 cytokine mRNA expression and mitogen-activated protein kinase signaling pathways in the splenic T cells of traumatized rats. Anesth Analg 2009; 109; 1666-73.

Wang SM, Kain ZN, White P. Acupuncture Analgesia: I. The Scientific Basis. Anesth Analg 2008a; 106; 602-10.

Wang SM, Kain ZN, White PF. Acupuncture analgesia: II. Clinical considerations. Anesth Analg 2008b; 106; 611-21, table of contents.

Wang Y, Zhang Y, Wang W, Cao Y, Han J-S. Effects of synchronous or asynchronous electroacupuncture stimulation with low versus high frequency on spinal opioid release and tail flick nociception. Experimental Neurology 2005; 192; 156-62.

Wen YR, Suter MR, Ji RR, Yeh GC, Wu YS, Wang KC, Kohno T, Sun WZ, Wang CC.

Activation of p38 mitogen-activated protein kinase in spinal microglia contributes to incision-induced mechanical allodynia. Anesthesiology 2009; 110; 155-65.

Wen YR, Tan PH, Cheng JK, Liu YC, Ji RR. Microglia: a promising target for treating neuropathic and postoperative pain, and morphine tolerance. J Formos Med Assoc 2011; 110; 487-94.

Wen YR, Wang CC, Yeh GC, Hsu SF, Huang YJ, Li YL, Sun WZ. DNIC-mediated analgesia produced by a supramaximal electrical or a high-dose formalin conditioning stimulus:

roles of opioid and alpha2-adrenergic receptors. J Biomed Sci 2010; 17; 19.

Wen YR, Yeh GC, Shyu BC, Ling QD, Wang KC, Chen TL, Sun WZ. A minimal stress model for the assessment of electroacupuncture analgesia in rats under halothane. Eur J Pain 2007; 11; 733-42.

Xu KD, Liang T, Wang K, Tian DA. Effect of pre-electroacupuncture on p38 and c-Fos expression in the spinal dorsal horn of rats suffering from visceral pain. Chin Med J (Engl) 2010; 123; 1176-81.

562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597

(32)

Xu WD, Zhu B, Rong PJ, Bei H, Gao XY, Li YQ. The pain-relieving effects induced by electroacupuncture with different intensities at homotopic and heterotopic acupoints in humans. Am J Chin Med 2003; 31; 791-802.

Yang J, Yang Y, Chen JM, Liu WY, Wang CH, Lin BC. Effect of oxytocin on acupuncture analgesia in the rat. Neuropeptides 2007; 41; 285-92.

Zeng YJ, Tsai SY, Chen KB, Hsu SF, Chen JY, Wen YR. Comparison of electroacupuncture and morphine-mediated analgesic patterns in a plantar incision-induced pain model.

Evid Based Complement Alternat Med 2014; 2014; 659343.

Zhang RX, Lao L, Wang L, Liu B, Wang X, Ren K, Berman BM. Involvement of opioid receptors in electroacupuncture-produced anti-hyperalgesia in rats with peripheral inflammation. Brain Res 2004; 1020; 12-7.

Zhang RX, Wang L, Liu B, Qiao JT, Ren K, Berman BM, Lao L. Mu opioid receptor- containing neurons mediate electroacupuncture-produced anti-hyperalgesia in rats with hind paw inflammation. Brain Res 2005a; 1048; 235-40.

Zhang RX, Wang L, Wang X, Ren K, Berman BM, Lao L. Electroacupuncture combined with MK-801 prolongs anti-hyperalgesia in rats with peripheral inflammation.

Pharmacol Biochem Behav 2005b; 81; 146-51.

Zhang Y, Li A, Lao L, Xin J, Ren K, Berman BM, Zhang RX. Rostral ventromedial medulla mu, but not kappa, opioid receptors are involved in electroacupuncture anti- hyperalgesia in an inflammatory pain rat model. Brain Res 2011; 1395; 38-45.

Zhuang ZY, Kawasaki Y, Tan PH, Wen YR, Huang J, Ji RR. Role of the CX3CR1/p38 MAPK pathway in spinal microglia for the development of neuropathic pain following nerve injury-induced cleavage of fractalkine. Brain Behav Immun 2007; 21;

642-51.

598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623

(33)

Figure Legends

Figure 1: Behavioral changes after plantar incision (PI). Behavioral changes measured by the

von Frey test (A) and Hargreave’s method (B) at different time points are shown. Heat withdrawal latencies returned to pre-incision baseline within one week, but mechanical withdrawal thresholds did not. Data are presented as mean ± SEM. ### p < 0.001 v.s. pre-PI baselines (BL). Repeated measurement ANOVA followed by post hoc Tukey’s test. N = 9/group.

Figure 2: Stimulation intensity affects analgesic effect of repeated EA at ST36 (Zusanli

acupoint) on PI-induced pain. After PI surgery, rats received daily EA of 3 mA (N = 9) or 10 mA (N = 8), respectively. The naïve group (N=7) did not have any surgery or EA treatment, and the sham group (N = 9) received PI and sham needling without electrical current. EA of 10 mA attenuated paw withdrawal responses to von Frey filaments (Fig. A), but not heat withdrawal latencies (Fig. B). Data are presented as mean ± SEM. * p < 0.05, *** p< 0.001 v.s. the PI+sham group; +++ p < 0.001 v.s. the PI+3 mA EA group, and ### p < 0.001 v.s. the other three groups; one-way ANOVA followed by post hoc Tukey’s test.

Figure 3: Repeated high-intensity EA enhances PI-induced p38 activation. The sham group received PI surgery and sham needling without electrical current. Immunostaining illustrates 624

625 626 627 628 629 630

631

632 633 634 635 636 637 638 639 640

641 642

(34)

p-p38 results in groups of the (A) PI+Sham, (B) PI+3 mA EA, and (C) PI+10 mA EA groups on Day 3. (D) Quantification of the p-p38-immunoreactive cells in the L4-5 spinal dorsal horn. N= 6/group. Data are presented as mean ± SEM. * p < 0.05 v.s. the PI+Sham group, one-way ANOVA followed by post hoc Tukey’s test. Scale bar = 50 μm.

Figure 4: PI-induced p-p38 is located within microglia in the spinal dorsal horn.

Immunofluorescent stainings of p-p38 (red, Fig. A, B, C), NeuN (A’, neuronal marker), OX- 42 (B’, microglia marker), and GFAP (C’, astrocytic marker) in the L4-5 spinal dorsal horn are shown. Double-staining demonstrates co-localization of p-p38 (red) in microglia (green) (B”, arrow heads), but not in neuron (A”) or astrocyte (C”). Scale bar = 25 μm.

Figure 5: Repeated EA stimulations induce p38 activation in the normal rats. Rats received

daily EA of 3 mA or 10 mA at right ST36 for 4 days. Immunostaining illustrates p-p38- immunoreative cells in groups of the (A) Naïve, (B) 3 mA EA, and (C) 10 mA EA. (D) Quantification of the p-p38-ir cells in the Naive group (N = 3), 3 mA EA (N = 4), and 10 mA EA (N = 4). Data are presented as mean ± SEM. *** p < 0.001 v.s. the Naïve group. One-way ANOVA followed by post hoc Tukey’s test. Scale bar = 50 μm.

643 644 645 646 647

648 649 650 651 652

653 654 655 656 657 658 659

660

(35)

Figure 6: p38 inhibitor, SB203580, decreases PI-induced mechanical allodynia, but not heat

hyperalgesia. (A) Intrathecal SB203580 dose-dependently prevents mechanical allodynia at 1 and 3 h after PI. The effect disappears on next day; (B) SB203580 has almost no effect on heat hyperalgesia at all time points. DMSO of 20% is the concentration of vehicle for maximal SB 203580 dose (2.0 mM). N = 8 for each group. Data are presented as mean ± SEM. * p < 0.05, ** p < 0.01, *** p < 0.001 v.s. the Saline group, and +++ < 0.001 v.s. the 0.2 mM SB group by one-way ANOVA followed by post hoc Tukey’s test.

Figure 7: Co-administration with low-dose SB203580 enhances analgesic effect of low-

intensity EA on PI-induced mechanical hypersensitivity. (A) Comparison of allodynic thresholds among the saline, the vehicle (2% DMSO), intrathecal low-dose SB203580 (0.2 mM SB), low-intensity EA (3-mA EA), and SB+EA (co-administration with 0.2-mM SB203580 and 3-mA EA) at different time points post PI. (B) Heat hyperalgesic responses among groups. N= 68 for each group. Data are presented as mean ± SEM. * p < 0.05, *** p

< 0.001 v.s. the Saline group, and +++ < 0.001 v.s. the SB+EA group by one-way ANOVA followed by post hoc Tukey’s test.

661 662 663 664 665 666 667 668

669 670 671 672 673 674 675 676

參考文獻

相關文件

CT images showed a low-density lesion in the periapical areas of the right mandible, which accompanied a well-de fined, high-density lesion, and a mixed low/high-density lesion with

In this talk, we present a novel algorithm by using adaptive grids as well as well as a tailored finite point method for a time dependent convection diffusion model to implement

In this paper, we provide new decidability and undecidability results for classes of linear hybrid systems, and we show that some algorithms for the analysis of timed automata can

In this section we define a general model that will encompass both register and variable automata and study its query evaluation problem over graphs. The model is essentially a

Recent preclinical data by Nardone et al (2015) indicate that olaparib may enhance endocrine therapy efficacy and circumvents resistance; as a consequence, addition of olaparib to

which can be used (i) to test specific assumptions about the distribution of speed and accuracy in a population of test takers and (ii) to iteratively build a structural

• We have found a plausible model in AdS/CFT that captures some essential features of the quantum hall eect as well as a possible experimental prediction. • Even if this is the

We showed that the BCDM is a unifying model in that conceptual instances could be mapped into instances of five existing bitemporal representational data models: a first normal