Graduate School of Acupuncture and Moxibustion, Meiji University of Oriental Medicine , Japan

Similar documents
Effect of varying frequency and duration of electroacupuncture stimulation on carrageenan-induced hyperalgesia

Analgesic roles of peripheral intrinsic met-enkephalin and dynorphin A in long-lasting inflammatory pain induced by complete Freund's adjuvant in rats

Neuroimmune mechanisms of opioid antinociception in early inflammation involvement of adhesion molecules

Receptors and Neurotransmitters: It Sounds Greek to Me. Agenda. What We Know About Pain 9/7/2012

NMDA-Receptor Antagonists and Opioid Receptor Interactions as Related to Analgesia and Tolerance

Repeated intra-articular injections of acidic saline produce long-lasting joint pain and. widespread hyperalgesia

TRAMADOL, A CENTRALLY ACTING OPIOID : ANTICONVULSANT EFFECT AGAINST MAXIMAL ELECTROSHOCK SEIZURE IN MICE

Acupuncture Painkiller Postoperative Narcotic Boost Found

WSVMA Annual Conference

Pharmacology of Pain Transmission and Modulation

Opioid Receptor Trafficking in Pain States

Ideal Parameters for Electroacupuncture Analgesia: A Review of Neurophysiological and Clinical Evidence

Leukocytes in the regulation of pain and analgesia

Naltrexone for the treatment of Crohn s disease

TRANSCUTANEOUS ELECTRICAL NERVE STIMULATION (TENS) Dr. Mohammed TA, Omar, PhD, PT Rehabilitation Science Department CAMS-KSU

Use of Percutaneous Electrical Nerve Stimulation (PENS) in the Short-term Management of Headache

Special Issue on Pain and Itch

Differential Regulation of Opioid Receptors During. Inflammation

Journal of Chemical and Pharmaceutical Research

GLOSSARY OF TERMS ASSOCIATED WITH TENS

CRITICALLY APPRAISED PAPER (CAP) Evidence / Title of article

Contribution of Opioid Receptors on Primary Afferent Versus Sympathetic Neurons to Peripheral Opioid Analgesia

PAIN IS A SUBJECTIVE EXPERIENCE: It is not a stimulus. MAJOR FEATURES OF THE PAIN EXPERIENCE: Sensory discriminative Affective (emotional) Cognitive

Accepted Manuscript S (16) Reference: PEP To appear in: Peptides

Pain. Pain. Pain: One definition. Pain: One definition. Pain: One definition. Pain: One definition. Psyc 2906: Sensation--Introduction 9/27/2006

Immune cell derived opioids protect against neuropathic pain in mice

PAIN & ANALGESIA. often accompanied by clinical depression. fibromyalgia, chronic fatigue, etc. COX 1, COX 2, and COX 3 (a variant of COX 1)

Research Article Effects of Electroacupuncture Treatment on Bone Cancer Pain Model with Morphine Tolerance

ONLINE. F.M. Teixeira*, L.L. Castro*, R.T. Ferreira, P.A. Pires, F.A. Vanderlinde and M.A. Medeiros

NADA Ear Acupuncture Protocol: Opportunities and Challenges for Integrating in Addiction and Behavioral Health Treatment

Opioidergic orofacial antinociception induced by electroacupuncture at acupoint St36

The Role of Peripheral Mu Opioid Receptors in the Modulation of Capsaicin-Induced Thermal Nociception in Rhesus Monkeys 1,2

VIVITROL (naltrexone for extended-release injectable suspension) A µ-opioid Receptor Antagonist

The Egyptian Journal of Hospital Medicine (January 2018) Vol. 70 (12), Page

By the end of this lecture the students will be able to:

Local administration of 9 -tetrahydrocannabinol attenuates capsaicin-induced thermal nociception in rhesus monkeys: a peripheral cannabinoid action

Electroacupuncture: Mechanisms and Clinical Application

Karam Darwish. Dr. Munir. Munir Gharaibeh

Pregnanolone effects on the blood pressure of stress-induced hypertension in rats

Published online October 10, 2016

Acupuncture Immune System Discovery

The Nervous System Mark Stanford, Ph.D.

Received: July 29, 2016 / Revised: July 29, 2016 / Accepted: August 23, 2016 c 2016 J Korean Soc Phys Med. Abstract 1)

Pain Pathways. Dr Sameer Gupta Consultant in Anaesthesia and Pain Management, NGH

Low-level laser therapy (LLLT) was pioneered in

San Francisco Chronicle, June 2001

Short communication NORADRENERGIC STIMULATION OF THE LATERAL HYPOTHALAMUS AS A REINFORCEMENT IN T MAZE LEARNING IN RATS

Review. How Do Acupuncture and Moxibustion Act? Focusing on the Progress in Japanese Acupuncture Research

SUPPLEMENTARY INFORMATION

Cholecystokinin antagonist, proglumide, stimulates growth hormone release in the rat

Somatosensory Physiology (Pain And Temperature) Richard M. Costanzo, Ph.D.

THE OPIUM POPPY OPIOID PHARMACOLOGY 2/18/16. PCTH 300/305 Andrew Horne, PhD MEDC 309. Papaver somniferum. Poppy Seeds Opiates

Medical Articles > Medical Article #6

Mechanism of action of Low Dose Naltrexone (LDN)

NEUROBIOLOGY ALCOHOLISM

Overactive Bladder; OAB. Aδ silent C. Adenosine triphosphate; ATP SD g n Key words

Biological and Behavioral Indicators of Stress in the Boar

MODULE 2 THE LABORATORY RAT

Acupuncture the scientific proof

SomeFacts... Joint pain and its treatment with acupuncture. Overview. Some Figures. Primary (Idiopathic) OA. Primary (Idiopathic) OA

Supplementary Figure 1

Neuroimmunology. Innervation of lymphoid organs. Neurotransmitters. Neuroendocrine hormones. Cytokines. Autoimmunity

Sensory Analgesia. Pain Definitions a distressing feeling due to disease, bodily injury or organic disorder. uneasiness of mind or grief.

Antidepressive-like effects of electroacupuncture in rats

Chapter 6 Communication, Integration, and Homeostasis

Acupoint Stimulation Using Bee Venom Attenuates Formalin-Induced Pain Behavior and Spinal Cord Fos Expression in Rats

No Tolerance to Peripheral Morphine Analgesia in Presence of Opioid Expression in Inflamed Synovia

Acupuncture for Well Being

Recent Advances in Energy, Environment, Biology and Ecology

Spinal Cord Injury Pain. Michael Massey, DO CentraCare Health St Cloud, MN 11/07/2018

Non-opioidergic mechanism mediating morphine-induced. antianalgesia in the mouse spinal cord. Hsiang-En Wu, Jonathan Thompson, Han-Sen Sun,

Central Progesterone Involvement in Estrogen- Induced Prolactin and Luteinizing Hormone Secretion Surges in Female Rats

Nervous System Communication. Nervous System Communication. The First Nerve Cells 1/2/11

CONTENTS. Disclosure of Conflict of Interest. None. Is opioid effective in long-term use? Can opioid exacerbate pain? Why are we still using opioids?

TRANSCUTANEOUS ELECTRICAL STIMULATION

Peripheral opioid receptor agonists for analgesia: A comprehensive review

Department of Mechanics and Engineering Science, Fudan University, Shanghai , China 2

University of Colorado-Boulder

Acupuncture for Well Being

VASILE HEFCO 1*, LUCIAN HRITCU 1, ADRIAN TIRON 1, ANDREEA-IOANA HEFCO 1

, , 2. ment of Physiology, Henan Medical U niversity, Zhengzhou

Slide 1. Slide 2. Slide 3. Opioid (Narcotic) Analgesics and Antagonists. Lesson 6.1. Lesson 6.1. Opioid (Narcotic) Analgesics and Antagonists

niap Terms and Definitions

significantly suppressed this increase in plasma ACTH.

INDUCTION OF OVULATION IN URETHANE-TREATED RATS

EFFECTS OF NARCOTIC AND NON-NARCOTIC ANALGESICS ON THE ABDOMINAL OR TAIL STIMULATION-INDUCED STRUGGLING IN RATS

Repeated stress exposure causes strain-dependent shifts in the behavioral economics of cocaine in rats

THE EFFECT OF THE CHOLINERGIC SYSTEM ON PLANTAR PAIN RESPONSE IN RATS

Choose a category. You will be given the answer. You must give the correct question. Click to begin.

OPTIMAL STIMULATION FREQUENCY OF TRANSCUTANEOUS ELECTRICAL NERVE STIMULATION ON PEOPLE WITH KNEE OSTEOARTHRITIS

At a Glance. Background Information. Lesson 3 Drugs Change the Way Neurons Communicate

International Conference on Biological Sciences and Technology (BST 2016)

What is Pain? An unpleasant sensory and emotional experience associated with actual or potential tissue damage. Pain is always subjective

The peripheral antinociceptive e ect induced by morphine is associated with ATP-sensitive K + channels

Pathophysiology of Pain

Analgesia for Patients with Substance Abuse Disorders. Lisa Jennings CN November 2015

Fibromyalgia: Current Trends and Concepts

A New Era of Migraine Management: The Challenging Landscape in Prevention

What do we want for pain medications?

MODULE 2 Substance Administration and Blood Collection in the Laboratory Rat

Transcription:

The American Journal of Chinese Medicine, Vol. 32, No. 2, 269 279 2004 World Scientific Publishing Company Institute for Advanced Research in Asian Science and Medicine Corticotropin-Releasing Factor and Interleukin-1β are Involved in the Electroacupuncture-Induced Analgesic Effect on Inflammatory Pain Elicited by Carrageenan Reina Sekido, * Keisou Ishimaru and Masakazu Sakita * * Departments of Surgery, and Clinical Acupuncture and Moxibustion Graduate School of Acupuncture and Moxibustion, Meiji University of Oriental Medicine 629-0392, Japan Abstract: Electroacupuncture (EA) is used to relieve various kinds of pain. However, the mechanistic basis of electroacupuncture analgesia (EAA) in inflammatory pain remains unclear. In the present study, we investigated whether endogenous peripheral corticotropin-releasing factor (CRF) or interleukin-1β (IL-1) participated in EAA during hyperalgesia elicited by carrageenan-induced inflammation. Carrageenan was subcutaneously administered by intraplantar (i.pl.) injection of the left hind paw to induce inflammation. Nociceptive thresholds were measured using the paw pressure threshold (PPT) (Randall Sellito Test). Rats received 3 Hz EA in the left anterior tibial muscles for 1 hour after carrageenan injection. The selective CRF antagonist, α-helical CRF, or the recombinant IL-1 receptor antagonist, IL-1ra, was administered by i.pl. injection of the inflamed paw or by intravenous (i.v.) injection 1 hour before EA. PPT decreased significantly 3 hours after carrageenan injection. This decrease persisted at least 24 hours after carrageenan injection. EA resulted in significant increases of PPT, moreover, PPT elevations lasted 24 hours after carrageenan injection. By contrast, PPT elevations produced by EA were dose-dependently antagonized by local i.pl. injection of α- helical CRF or IL-1ra. This PPT elevation was not influenced by i.v. injection of α-helical CRF or IL-1ra. These findings suggest that peripheral CRF or IL-1 participate in EAA during hyperalgesia. The release of CRF or IL-1 elicited by EA may trigger the release of opioid peptides within inflamed tissue which may activate peripheral opioid receptors and inhibit the pain. Keywords: Electroacupuncture; Analgesia; Corticotropin-releasing Factor; IL-1β; Carrageenan; Hyperalgesia; Inflammation. Correspondence to: Dr. Reina Sekido, Department of Surgery, Graduate School of Acupuncture and Moxibustion, Meiji University of Oriental Medicine, Hiyoshi, Funai, Kyoto, 629-0392, Japan. Tel: (+81) 771-72-1181, Fax: (+81) 771-72-0326, E-mail: r_sekido@muom.meiji-u.ac.jp. 269

270 R. SEKIDO et al. Introduction Pain, evoked by local injury and inflammation, can be controlled by various endogenous mechanisms. Under inflammatory conditions, opioid-containing immune cells migrate preferentially to inflamed sites (Przewlocki et al., 1992; Stein, 1995; Cabot et al., 1997). Experimental and clinical studies demonstrate that locally administered opioid receptor agonists can produce pronounced analgesic effects by interacting with peripheral opioid receptors in inflamed tissue (Antonijevic et al., 1995; Stein et al., 1989, 1990a, 1993 and 1995). Stein et al. (1990a and b) have also shown that environmental stimuli, such as in the cold water swim (CWS) in rats with unilateral hind paw inflammation elicit peripheral opioid receptor-mediated anti-nociception in the inflamed paw. These opioids are released locally from immunocytes during environmental stimuli and inhibit pain in animals (Stein et al., 1990a and b) and in humans (Stein et al., 1993). Moreover, these opioid peptides can be released by stimulating immunocytes with corticotropin-releasing factor (CRF) or interleukin- 1β (IL-1) in vivo and in vitro (Schafer et al., 1994; Cabot et al., 2001). It has also been shown that the anti-nociception produced by CWS is blocked by intraplantar (i.pl.) injection of CRF antagonist, suggesting a peripheral receptor-specific action of endogenous CRF (Schafer et al., 1996). Thus, it is believed that local immune system and peripheral opioid receptors are involved in inflammatory pain control (Stein et al., 1993; Stein, 1995). At present, electroacupuncture (EA) is used to relieve various kinds of pain. However, little is known about the effects of electroacupuncture analgesia (EAA) on inflammatory pain. In a previous study, we observed that peripheral opioid receptors might be involved in the EAA in inflamed tissue (Sekido et al., 2000). In this study, we investigated whether peripheral CRF or IL-1 might be the endogenous trigger for local opioid release and was involved in EAA in rats with carrageenan-induced inflammation. Materials and Methods Animals Male Sprague-Dawley rats weighing 280 380 g were purchased from Japan SLC, Inc. The animals were kept at 24 ± 1ºC and a relative humidity of 50% to 60%. Standard laboratory rodent food and tap water were available ad libitum. All experiments were conducted in the light phase of a 12/12-hour (7 am/7 pm) light-dark cycle. This study followed NIH regulations for humane experimentation on animals and the guidelines of the International Association for the Study of Pain. Animals were treated in accordance with the guidelines of our Institutional Committee on the Treatment of Experimental Animals. Induction of Inflammation Carrageenan (2%, 0.1 ml; Sigma, St. Louis, MO) was subcutaneously administered by i.pl. injection of the left hind paw of rats under ether anesthesia using a 26-gauge needle to induce inflammation.

CRF AND IL-1β ON ACUPUNCTURE ANALGESIA 271 Experimental Protocols To determine the involvement of peripheral CRF or IL-1 on local opioid release, we examined whether α-helical CRF or recombinant IL-1 receptor antagonist (IL-1ra) given by i.pl. injection to the inflamed paw blocked the paw pressure threshold (PPT) elevations produced by EA. Animals received i.pl. injection (0.1 ml) of α-helical CRF (2.5, 3.7 and 5.0 ng) or IL-1ra (25, 50 and 100 ng) 1 hour before EA (n = 6 per group). To confirm that these effects were not mediated through a central site of action, another group of animals received an intravenous (i.v.) injection (0.1 ml) of 5.0 ng α-helical CRF or 100 ng IL-1ra 1 hour before EA (n = 6 per group). Algesiometry Nociceptive thresholds were evaluated using an Analgesy-meter (Ugo Basile). Rats were gently restrained under a soft cloth jacket and incremental pressure was applied onto the dorsal surface of the left hind paw. The pressure required to elicit paw withdrawal, the PPT, was determined. We used a cut-off threshold of 250 g to avoid tissue damage to the paw. The mean of two consecutive measurements, separated by 2 minutes, was determined after a rest period of 15 minutes. PPT was determined 15 minutes before, just before, and 3, 4, 5, 7, 9 and 24 hours after the carrageenan injection. Electroacupuncture A pair of stainless steel needles, 0.20 mm in diameter and 30 mm in length, were inserted into an acupoint of Zusanli (ST36) and the left anterior tibial muscles (5 mm from the Zusanli). A 3 Hz biphasic square wave pulse of 0.1 ms pulse width was delivered via the needles for a period of 60 minutes using a constant current programmed pulse generator. EA was started from 3 hours after carrageenan injection. The intensity of EA was increased according to a schedule of 1-2-3 ma, for 20 minutes at each intensity. The intensity was sufficient to produce a rhythmic muscle contraction of the hind legs. The rats were gently restrained under a soft cloth jacket during the PPT measurement and EA procedure. Except at these times, rats were left in their cage to move freely. Drugs We used the selective CRF antagonist α-helical CRF (Sigma) and recombinant IL-1 receptor antagonist IL-1ra (R&D Systems). Both agents were dissolved in sterile distilled water and administered by i.pl. injection (0.1 ml) of the inflamed paw or i.v. injection via a tail vein 1 hour before EA. Control animals received 0.1 ml of sterile distilled water.

272 R. SEKIDO et al. Data Analysis The experimental data were expressed as means ± SD. Repeated measures ANOVA were performed to determine the overall effect. Tukey s post-hoc test was then used to determine probability values when repeated measure ANOVAs indicated a significant effect. p < 0.05 was considered as statistically significant. Results Effect of EA during Hyperalgesia Elicited by Carrageenan-induced Inflammation Figure 1 shows the time course of analgesia produced by EA during inflammatory hyperalgesia. In the control group, PPT just before carrageenan injection was 83.9 ± 13.4 g. Three hours after the carrageenan injection, a marked ipsilateral inflammatory response (swelling and redness) appeared and PPT decreased significantly (54.1 ± 14.2 g). Moreover, this decrease continued for 24 hours after carrageenan injection. In the EA group, PPT decreased to almost the same level as the control group 3 hours after the carrageenan injection. However, PPT increased significantly (86.4 ± 12.3 g) immediately after the EA (p < 0.001). The PPT elevations produced by EA lasted at least 20 hours after the EA (24 hour carrageenan injection) (Fig. 1). paw pressure threshold (g) 120 * # 100 80 60 40 20 ~ carrageenan EA -15min 0 3 4 5 7 9 24 * h cont EA Figure 1. Effects of EA during hyperalgesia elicited by carrageenan-induced inflammation. The results are expressed as mean ± SD. n = 6 per group, * p < 0.001, # p < 0.01, p < 0.05 (control versus EA).

CRF AND IL-1β ON ACUPUNCTURE ANALGESIA 273 Effect of i.pl. Injection of α-helical CRF on EAA during Hyperalgesia Elicited by Carrageenan-induced Inflammation Figure 2A shows the effect of i.pl. injection of α-helical CRF on EAA during inflammatory hyperalgesia. To observe the effect of α-helical CRF on the PPT elevations induced by EA, rats were given an i.pl. injection of α-helical CRF (2.5, 3.7 and 5.0 ng) or vehicle 1 hour before EA. In the α-helical CRF (2.5, 3.7 and 5.0 ng) + EA or vehicle + EA groups, PPT decreased to almost the same level as the control group 3 hours after the carrageenan injection. In the vehicle + EA group, PPT significantly increased immediately, 5 hours and 20 hours after termination of EA (89.1 ± 10.5 g, 77.7 ± 10.4 g and 78.5 ± 6.4 g, respectively), compared to that of the control group. In the 2.5 ng α-helical CRF + EA group, PPT increased to the same level as the vehicle + EA group after the termination of EA, and no differences were observed between these two groups. In the 3.7 ng α-helical CRF + EA group, PPT tended to increase immediately after the termination of EA and thereafter compared to that of the control group. In the 5.0 ng α-helical CRF + EA group, however, PPT did not increase immediately, 5 hours and 20 hours after the termination of EA (55 ± 6.1 g, 53.7 ± 2.6 g and 48.7 ± 4.1 g, respectively) and no differences were observed between this group and the control group, but significant differences were observed compared to that of the vehicle + EA group. PPT elevations produced by EA were not influenced by i.v. injection of 5.0 ng α-helical CRF and PPT significantly increased during 24 hours after the carrageenan injection compared to that of the control group (Fig. 2B). Effect of i.pl. Injection of IL-1ra on EAA during Hyperalgesia Elicited by Carrageenan-induced Inflammation Figure 3A shows the effect of i.pl. injection of IL-1ra on EAA during inflammatory hyperalgesia. Rats were given an i.pl. injection of 25, 50 and 100 ng IL-1ra 1 hour before EA. In the IL-1ra (25, 50 and 100 ng) + EA groups, PPT decreased to the same level as the control group 3 hours after carrageenan injection. In the 25 ng IL-1ra + EA group, PPT increased to the same level as the vehicle + EA group immediately, 1 hour and 5 hours after the termination of EA (86.8 ± 13.3 g, 81.0 ± 7.9 g and 80.4 ± 9.6 g, respectively). In the 50 ng IL-1ra + EA group, PPT tended to increase after the termination of EA compared to that of the control group. In the 100 ng IL-1ra + EA group, however, PPT did not increase after the termination of EA. Thus, PPT elevations elicited by EA were dose-dependently antagonized by i.pl., but not by i.v. injection of α-helical CRF or IL-1ra (Figs. 2 and 3). In addition, PPT elevations elicited by EA were not antagonized by i.v. injection of 100 ng IL-1ra (Fig. 3B). Moreover, neither i.pl. injection of α-helical CRF nor IL-1ra per se did not have analgesic or hyperalgesic effects during carrageenan-induced inflammation (Table 1).

274 R. SEKIDO et al. paw pressure threshold (g) 120 A 100 80 60 40 0 ~ carrageenan EA * * * * * * * Control Vehicle + EA 2.5 ng + EA 3.7 ng + EA 5.0 ng + EA paw pressure threshold (g) 120 B * 100 80 60 40 ~ carrageenan 0-15min EA # 0 3 4 5 7 9 24 h Control 5.0 ng i.v. + EA Figure 2. Effects of i.pl. (A) or i.v. (B) injection of α-helical CRF on EAA during hyperalgesia elicited by carrageenan-induced inflammation. The results are expressed as mean ± SD. n = 6 per group, * p < 0.001, # p < 0.01, p < 0.05 (control versus vehicle + EA, vehicle + EA versus 2.5 ng + EA, vehicle + EA versus 5.0 ng + EA, control versus 5.0 ng i.v.+ EA).

CRF AND IL-1β ON ACUPUNCTURE ANALGESIA 275 paw pressure threshold (g) paw pressure threshold (g) 120 100 80 60 40 0 120 100 80 60 40 A ~ carrageenan B ~ carrageenan 0-15min EA EA * * # # # * * * * * # 0 3 4 5 7 9 24 Figure 3. Effects of i.pl. (A) or i.v. (B) injection of IL-1ra on EAA during hyperalgesia elicited by carrageenaninduced inflammation. The results are expressed as mean ± SD. n = 6 per group, * p < 0.001, # p < 0.01, p < 0.05 (control versus vehicle + EA, vehicle + EA versus 25 ng + EA, vehicle + EA versus 100 ng + EA, vehicle + EA versus 50 ng + EA, control versus 100 ng i.v. + EA). * Control Vehicle + EA 25 ng + EA 50 ng + EA 100 ng + EA Control 100 ng i.v. + EA h

276 Table 1. Effects of i.pl. Injection of α-helical CRF or IL-1ra During Hyperalgesia Elicited by Carrageenan-induced Inflammation Group 15 Minutes 0 Hours 3 Hours 4 Hours 5 Hours 7 Hours 9 Hours 24 Hours Carrageenan control 86.2 ± 15.3 83.9 ± 13.4 54.1 ± 14.2 54.1 ± 9.2 60.0 ± 16.6 58.7 ± 20.3 56.4 ± 9.4 57.5 ± 8.2 (n = 6) (100) (97.3) (62.8) (62.8) (69.5) (68.1) (65.4) (66.6) Carrageenan + 73.3 ± 11.6 72.0 ± 10.1 58.7 ± 11.4 43.7 ± 9.7 43.7 ± 5.0 49.5 ± 6.8 47.9 ± 10.6 50.8 ± 7.3 α-helical CRF 2.5 ng (100) (98.2) (80.1) (59.6) (59.6) (67.6) (65.3) (69.3) (n = 3) Carrageenan + 74.5 ± 8.7 72.0 ± 10.0 55.0 ± 4.5 53.3 ± 4.3 53.3 ± 6.2 50.4 ± 5.7 50.8 ± 5.0 58.3 ± 8.5 α-helical CRF 3.7 ng (100) (96.6) (73.7) (71.5) (71.5) (67.5) (68.1) (78.2) (n = 3) Carrageenan + 77.5 ± 19.2 77.9 ± 22.5 53.7 ± 9.9 51.2 ± 8.7 50.8 ± 8.3 48.7 ± 3.3 55.0 ± 2.5 60.4 ± 4.7 α-helical CRF 5.0 ng (100) (100.5) (69.3) (66.1) (65.5) (62.9) (70.9) (77.9) (n = 3) Carrageenan + 65.4 ± 4.0 64.1 ± 1.9 46.6 ± 8.1 45.4 ± 0.7 42.5 ± 1.2 45.8 ± 8.0 48.3 ± 5.0 51.6 ± 5.2 IL-1ra 25 ng (100) (98.0) (71.3) (69.4) (64.9) (70.0) (73.8) (78.9) (n = 3) Carrageenan + 78.7 ± 9.1 75.9 ± 6.8 55.9 ± 2.5 52.1 ± 4.8 46.8 ± 5.9 53.7 ± 6.5 52.1 ± 4.6 55.6 ± 5.2 IL-1ra 50 ng (100) (96.4) (71.0) (66.2) (59.5) (68.2) (66.2) (70.6) (n = 4) Carrageenan + 75.9 ± 15.8 78.4 ± 18.7 57.8 ± 12.1 57.1 ± 11.1 50.0 ± 5.3 57.0 ± 16.7 55.3 ± 9.8 63.1 ± 10.2 IL-1ra 100 ng (100) (103.2) (76.1) (75.3) (65.8) (75.1) (72.8) (83.1) (n = 4) The results are expressed as mean ± SD (g) and as percentage change from PPT of 15 minutes before carrageenan injection (100%). n = 6; control, n = 3 4 per group. R. SEKIDO et al.

CRF AND IL-1β ON ACUPUNCTURE ANALGESIA 277 Discussion EA has been widely used to relieve various kinds of pain. Numerous investigations of the mechanism underlying EAA have been performed in humans and animals. The results have shown that EAA was reversed by naloxone, an opioid receptor antagonist (Mayer et al., 1977; He, 1987; Chen et al., 1996), and that the quantity of β-endorphin or enkephalin in the cerebrospinal fluid was increased after EA (Clement-Jones et al., 1979 and 1980; He, 1987). Although it is well known that EAA is at least partially mediated by endogenous opioids and other neurotransmitters in the central nervous system (CNS) (Han and Trenius, 1982; He, 1987), the mechanism underlying EAA during inflammatory pain is unclear. In a previous study, we showed that the EAA during inflammatory hyperalgesia lasted longer and was blocked by i.pl. injection of naloxone (Sekido et al., 2000). This indicated that peripheral opioid receptors might be involved in the EAA during inflammatory hyperalgesia. We then investigated whether the endogenous peripheral CRF or IL-1 participated in EAA during hyperalgesia elicited by carrageenan-induced inflammation. In the present study, we found that the EAA in inflamed tissue could be dose-dependently blocked by local i.pl. injection of α-helical CRF or IL-1ra. This phenomenon is in accordance with the finding that anti-nociception elicited by CWS is blocked by i.pl. injection of α-helical CRF (Schafer et al., 1996). CRF and IL-1 receptors have been demonstrated to be expressed in splenocytes, macrophages, and T- and B-lymphocytes (Webster et al., 1990; Dinarello and Thompson, 1991; Karalis et al., 1991; Crofford et al., 1992; Mousa et al., 1996) and these receptors are upregulated in inflamed tissue (Mousa et al., 1996). CRF and IL-1 receptors are also detectable in the CNS (Heijnen et al., 1991). However, peripheral CRF and IL-1 receptors on immunocytes were probably involved in the EAA since the EAA in inflamed tissue could not be blocked by i.v. injection of α-helical CRF or IL-1ra. There are several differences between CWS-induced anti-nociception and EAA. First, EAA in inflamed tissue lasted 20 hours after termination of EA while the CWS-induced anti-nociception returned to baseline 10 17 minutes after termination of CWS (Schafer et al., 1996). Secondly, EAA was blocked by i.pl. injection of both α-helical CRF or IL-1ra. However, CWS-induced anti-nociception could not be blocked by i.pl. injection of IL-1ra (Schafer et al., 1996). These differences might be due to the difference in inflammatory agent used and stage of inflammatory reaction. Stein et al. used Freund s complete adjuvant to induce inflammation and CWS was conducted 4 days after inoculation. By contrast, we used carrageenan to induce inflammation and EA was conducted 3 hours after carrageenan injection. Antonijevic et al. (1995) showed that inflammation-induced disruption of the perineurial barrier and peripheral opioid analgesia coincides with earlier stages of an inflammatory reaction. In this study, disruption of the perineurial barrier might be involved in EAA. IL-1 gene expression has also been shown to peak very early, i.e. 1 2 hours after inflammatory stimuli (Ulich et al., 1992). Therefore, the EAA might be antagonized by i.pl. injection of IL-1ra. The other possible difference between EAA and CWS-induced anti-nociception is in the mechanism of action. It is well known that the analgesic effect of EA is mediated by

278 R. SEKIDO et al. endogenous opioids and other neurotransmitters including serotonin and norepinephrine in the CNS (Han and Trenius, 1982). Therefore, we could not completely exclude the involvement of the CNS in the mechanism of EAA in rats with carrageenan-induced inflammation. Taken together, during hyperalgesia elicited by carrageenan-induced inflammation, EA might be able to release CRF and IL-1 from immunocytes within inflamed tissue. CRF or IL-1 triggers the release of opioid peptides within inflamed tissue and these peptides activate peripheral opioid receptors, reducing neuronal excitability or release of proinflammatory neuropeptides (e.g. substance P), thereby inhibiting pain. Thus, in addition to the endogenous opioid mechanism in the CNS, the local immune system may be involved in the EAA. Further study is needed to clarify the mechanism of EAA during peripheral inflammation. References Antonijevic, I., S.A. Mousa, M. Schäfer and C. Stein. Perineurial defect and peripheral opioid analgesia in inflammation. J. Neurosci. 15: 165 172, 1995. Cabot, P.J., L. Carter, M. Schäfer and C. Stein. Methionine- enkephalin- and dynorphin A-release from immune cells and control of inflammatory pain. Pain 93: 207 212, 2001. Cabot, P.J., L. Carter, C. Gaiddon, Q. Zhang, M. Schäfer, J.P. Loeffler and C. Stein. Immune cellderived β-endorphin: production, release and control of inflammatory pain in rats. J. Clin. Invest. 100: 142 148, 1997. Chen, X.H., E.B. Geller and M.W. Adler. Electrical stimulation at traditional acupuncture sites in periphery produces brain opioid-receptor-mediated antinociception in rats. J. Pharmacol. Exp. Ther. 277: 654 660, 1996. Clement-Jones, V., P.J. Lowry, L. McLoughlin, G.M. Besser, L.H. Rees and H.L. Wen. Acupuncture in heroin addicts: changes in met-enkephalin and beta-endorphin in blood and CSF. Lancet ii: 380 383, 1979. Clement-Jones, V., L. McLoughlin, S. Tomlin, G.M. Besser, L.H. Rees and H.L. Wen. Increased betaendorphin but not met-enkephalin levels in human CSF after acupuncture for recurrent pain. Lancet ii: 946 949, 1980. Crofford, L.J., H. Sano, K. Karalis, E.L. Webster, E.A. Goldmuntz, G.P. Chrousos and R.L. Wilder. Local secretion of corticotropin-releasing hormone in the joints of Lewis rats with inflammatory arthritis. J. Clin. Invest. 90: 2555 2564, 1992. Dinarello, C.A. and R.C. Thompson. Blocking IL-1: interleukin-1 receptor antagonist in vivo and in vitro. Immunol. Today 12: 404 410, 1991. Han, J.S. and L. Trenius. Neurochemical basis acupuncture analgesia. Annu. Rev. Pharmacol. Toxicol. 22: 193 220, 1982. He, L.F. Involvement of endogenous opioid peptides in acupuncture analgesia. Pain 31: 99 121, 1987. Heijnen, C.J., A. Kavelaars and R.E. Ballieux. β-endorphin: cytokine and neuropeptide. Immunol. Rev. 119: 41 63, 1991. Karalis, K., H. Sano, J. Redwine, S. Listwak, R.L. Wilder and G.P. Chrousos. Autocrine or paracrine inflammatory actions of corticotropin-releasing hormone in vivo. Science 254: 421 423, 1991. Mayer, D.J., D.D. Price and A. Rafii. Antagonism of acupuncture analgesia in man by the narcotic antagonist naloxone. Brain Res. 121: 368 372, 1977.

CRF AND IL-1β ON ACUPUNCTURE ANALGESIA 279 Mousa, S.A., M. Schäfer, W.M. Mitchell, A.H.S. Hassan and C. Stein. Local upregulation of corticotropin-releasing hormone and interleukin-1 receptors in rats with painful hindlimb inflammation. Eur. J. Pharamacol. 311: 221 231, 1996. Przewlocki, R., A.H.S. Hassan, W. Lason, C. Epplen, A. Herz and C. Stein. Gene expression and localization of opioid peptides in immune cells of inflamed tissue: functional role in antinociception. Neuroscience 48: 491 500, 1992. Schäfer, M., L. Carter and C. Stein. Interleukin-1β and corticotropin-releasing factor inhibit pain by releasing opioids from immune cells in inflamed tissue. Proc. Natl. Acad. Sci. USA 91: 4219 4223, 1994. Schäfer, M., S.A. Mousa, Q. Zhang, L. Carter and C. Stein. Expression of corticotropin-releasing factor in inflamed tissue is required for intrinsic peripheral opioid analgesia. Proc. Natl. Acad. Sci. USA 93: 6096 6100, 1996. Sekido, R., K. Ishimaru and M. Sakita. Effect of electroacupuncture stimulation on hyperalgesia by carrageenan-induced inflammation. Bull. Meiji. Univ. Orient. Med. 26: 33 41, 2000. Stein, C. The control of pain in peripheral tissue by opioids. N. Engl. J. Med. 332: 1685 1690, 1995. Stein, C., A.H.S. Hassan, K. Lehrberger, J. Giefing and A. Yassouridis. Local analgesic effect of endogenous opioid peptides. Lancet 342: 321 324, 1993. Stein, C., C. Gramsch and A. Herz. Intrinsic mechanisms of antinociception inflammation: local opioid receptors and beta-endorphin. J. Neurosci. 10: 1292 1298, 1990a. Stein, C., A.H.S. Hassan, R. Przewlocki, C. Gramsch, K. Peter and A. Herz. Opioids from immunocytes interact with receptors on sensory nerves to inhibit nociception in inflammation. Proc. Natl. Acad. Sci. USA 87: 5935 5939, 1990b. Stein, C., M.J. Millan, T.S. Shippenberg, K. Peter and A. Herz. Peripheral opioid receptors mediating antinociception in inflammation. Evidence for involvement of mu, delta and kappa receptors. J. Pharmacol. Exp. Ther. 248(3): 1269 1275, 1989. Ulich, T.R., K. Guo, S. Yin, J. del Castillo, E.S. Yi, R.C. Thompson and S.P. Eisenberg. Endotoxininduced cytokine gene expression in vivo. IV. Expression of interleukin-1 alpha/beta and interleukin-1 receptor antagonist mrna during endotoxemia and during endotoxin-initiated local acute inflammation. Am. J. Pathol. 141: 61 68, 1992. Webster, E.L., D.E. Trancey, M.A. Jutila, S.A. Wolfe and E.B. De Souza. Corticotropin-releasing factor receptors in mouse spleen: identification of receptors-bearing cells as resident macrophages. Endocrinology 127: 440 452, 1990.