• Title/Summary/Keyword: Pain%3A hyperalgesia

Search Result 62, Processing Time 0.033 seconds

Muscimol as a treatment for nerve injury-related neuropathic pain: a systematic review and meta-analysis of preclinical studies

  • Hamzah Adel Ramawad;Parsa Paridari;Sajjad Jabermoradi;Pantea Gharin;Amirmohammad Toloui;Saeed Safari;Mahmoud Yousefifard
    • The Korean Journal of Pain
    • /
    • v.36 no.4
    • /
    • pp.425-440
    • /
    • 2023
  • Background: Muscimol's quick onset and GABAergic properties make it a promising candidate for the treatment of pain. This systematic review and meta-analysis of preclinical studies aimed at summarizing the evidence regarding the efficacy of muscimol administration in the amelioration of nerve injury-related neuropathic pain. Methods: Two independent researchers performed the screening process in Medline, Embase, Scopus and Web of Science extracting data were extracted into a checklist designed according to the PRISMA guideline. A standardized mean difference (SMD [95% confidence interval]) was calculated for each. To assess the heterogeneity between studies, 2 and chi-square tests were utilized. In the case of heterogeneity, meta-regression and subgroup analyses were performed to identify the potential source. Results: Twenty-two articles met the inclusion criteria. Pooled data analysis showed that the administration of muscimol during the peak effect causes a significant reduction in mechanical allodynia (SMD = 1.78 [1.45-2.11]; P < 0.0001; I2 = 72.70%), mechanical hyperalgesia (SMD = 1.62 [1.28-1.96]; P < 0.0001; I2 = 40.66%), and thermal hyperalgesia (SMD = 2.59 [1.79-3.39]; P < 0.0001; I2 = 80.33%). This significant amendment of pain was observed at a declining rate from 15 minutes to at least 180 minutes post-treatment in mechanical allodynia and mechanical hyperalgesia, and up to 30 minutes in thermal hyperalgesia (P < 0 .0001). Conclusions: Muscimol is effective in the amelioration of mechanical allodynia, mechanical hyperalgesia, and thermal hyperalgesia, exerting its analgesic effects 15 minutes after administration for up to at least 3 hours.

Therapeutic Effect of Epidurally Administered Lipo-Prostaglandin E1 Agonist in a Rat Spinal Stenosis Model

  • Park, Sang Hyun;Lee, Pyung Bok;Choe, Ghee Young;Moon, Jee Yeon;Nahm, Francis Sahngun;Kim, Yong Chul
    • The Korean Journal of Pain
    • /
    • v.27 no.3
    • /
    • pp.219-228
    • /
    • 2014
  • Background: A lipo-prostaglandin E1 agonist is effective for the treatment of neurological symptoms of spinal stenosis when administered by an oral or intravenous route. we would like to reveal the therapeutic effect of an epidural injection of lipo-prostaglandin E1 on hyperalgesia in foraminal stenosis. Methods: A total of 40 male Sprague-Dawley rats were included. A small stainless steel rod was inserted into the L5/L6 intervertebral foramen to produce intervertebral foraminal stenosis and chronic compression of the dorsal root ganglia (DRG). The rats were divided into three groups: epidural PGE1 (EP) (n = 15), saline (n = 15), and control (n = 10). In the EP group, $0.15{\mu}g{\cdot}kg-1$ of a lipo-PGE1 agonist was injected daily via an epidural catheter for 10 days from postoperative day 3. In the saline group, saline was injected. Behavioral tests for mechanical hyperalgesia were performed for 3 weeks. Then, the target DRG was analyzed for the degree of chromatolysis, chronic inflammation, and fibrosis in light microscopic images. Results: From the fifth day after lipo-PGE1 agonist injection, the EP group showed significant recovery from mechanical hyperalgesia, which was maintained for 3 weeks (P < 0.05). Microscopic analysis showed much less chromatolysis in the EP group than in the saline or control groups. Conclusions: An epidurally administered lipo-PGE1 agonist relieved neuropathic pain, such as mechanical hyperalgesia, in a rat foraminal stenosis model, with decreasing chromatolysis in target DRG. We suggest that epidurally administered lipo-PGE1 may be a useful therapeutic candidate for patients with spinal stenosis.

Inflammatory cytokines in midbrain periaqueductal gray contribute to diabetic induced pain hypersensitivity through phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signaling pathway

  • Guo, Mochi;Jiang, Zongming;Chen, Yonghao;Wang, Fei;Wang, Zhifeng
    • The Korean Journal of Pain
    • /
    • v.34 no.2
    • /
    • pp.176-184
    • /
    • 2021
  • Background: Diabetes-related neuropathic pain frequently occurs, and the underpinning mechanism remains elusive. The periaqueductal gray (PAG) exhibits descending inhibitory effects on central pain transmission. The current work aimed to examine whether inflammatory cytokines regulate mechanical allodynia and thermal hyperalgesia induced by diabetes through the phosphoinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) pathway in the PAG. Methods: Streptozotocin (STZ) was administered intraperitoneally to mimic allodynia and hyperalgesia evoked by diabetes in rats. Behavioral assays were carried out for determining mechanical pain and thermal hypersensitivity. Immunoblot and ELISA were performed to examine PAG protein amounts of interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α), as well as their corresponding receptors in STZ rats, and the expression of PI3K/protein kinase B (Akt)/mTOR signaling effectors. Results: Increased PAG p-PI3K/p-Akt/p-mTOR protein amounts were observed in STZ-induced animals, a PI3K-mTOR pathway inhibition in the PAG attenuated neuropathic pain responses. Moreover, the PAG concentrations of IL-1β, IL-6, and TNF-α and their receptors (namely, IL-1R, IL-6R, and tumor necrosis factor receptor [TNFR] subtype TNFR1, respectively) were increased in the STZ rats. Additionally, inhibiting IL-1R, IL-6R, and TNFR1 ameliorated mechanical allodynia and thermal hyperalgesia in STZ rats, alongside the downregulation of PI3K-mTOR signaling. Conclusions: Overall, the current study suggests that upregulated proinflammatory cytokines and their receptors in the PAG activate PI3K-mTOR signaling, thereby producing a de-inhibition effect on descending pathways in modulating pain transmission, and eventually contributing to neuropathic pain.

Effects of Ketamine on the Causalgiform Pain Produced by the Tight Ligation Of L5, L6 Spinal Nerves in the Rat (백서에서 제 5, 6번 요추신경의 결찰에 의하여 유발된 작열통성 통증에 대한 Ketamine의 치료효과)

  • Lee, Won-Hyung;Han, Neung-Hee;Kim, Tae-Heon
    • The Korean Journal of Pain
    • /
    • v.8 no.1
    • /
    • pp.18-24
    • /
    • 1995
  • We produced the causalgiform pain by the tight ligation of L5 and L6 spinal nerves in the adult rats. To evalute the effect of Ketamine -noncompetitive NMDA (N-methyl-D aspartate) antagoinst- on the causalgiform pain, we tested the changes of; withdrawal sensitivity to the innocuous mechanical stimulation of Von Frey hair 2.35 g(mechanical allodynia); withdrawal frequency to the cold stimulation of acetone (cold allodynia); and total withdrawal time (second) to the cold ($4^{\circ}C$) plate stimulation (cold hyperalgesia) after the administration of 1 mg, 3 mg, 10 mg/kg ketamine. The results were as follows: 1) Cold hyperalgesia was significantly reduced (p<0.05) by 1 mg, 3mg, 01 mg/kg ketamine. 2) Cold allodynia and mechanical allodynia was significantly reduced (p<0.05) by 10 mg/kg ketamine. Above results suggest a therapeutic utility of ketamine in treatment of causalgia - especially, cold hyperalgesia.

  • PDF

Carrageenan-Induced Hyperalgesia Is Partially Alleviated by Endomorphin-1 Locally Delivered into Inflamed Paws in Rat

  • Lee, Seo-Eun;Shin, Hong-Kee
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.7 no.6
    • /
    • pp.369-373
    • /
    • 2003
  • This study was performed to test whether endomorphin-1 has analgesic effect, when locally administrated into inflamed peripheral tissue. Carrageenan suspension (0.5%) was injected intraplantarly into the right paw of Sprague-Dawley male rats, and the rats were subjected to a series of mechanical stimuli with von Frei filaments before and after the injection. Carrageenan-injected rats showed typical inflammatory hyperalgesic signs and decrease of withdrawal threshold, peaked at 3 to 6 hours after the injection and lasted more than 3 days. Endomorphin-1 was intraplantarly injected with carrageenan, simultaneously or 3∼4 hours after carrageenan. Simultaneous injection of endomorphin-1 with carrageenan significantly reduced hyperalgesia and thd analgesic effect was prolonged up to 8 hours. The delivery of endomorphin-1 ($50{\mu}g$) into the inflamed area after 3 to 4 hours of carrageenan injection significantly increased the threshold of hyperalgesic mechanical withdrawal response, but only partially. Intrathecal treatment of endomorphin-1 completely reversed carrageenan-induced hyperalgesia. This report is the first to show that peripherally delivered endomorphin-1 relieved inflammatory hyperalgesia. But a control through peripheral ${\mu}-opioid$ receptors appears to be not sufficient for complete pain treatment.

Mechanism of Hyperalgesia Following Cutaneous Inflammation by Complete Freund Adjuvant (Complete Freund Adjuvant에 의한 피부염증에서 통각과민현상의 기전)

  • Jeong, Yong;Leem, Joong-Woo;Chung, Seung-Soo;Kim, Yun-Suk;Yoon, Duck-Mi;Nam, Taick-Sang;Paik, Kwang-Se
    • The Korean Journal of Pain
    • /
    • v.13 no.2
    • /
    • pp.164-174
    • /
    • 2000
  • Background: After an injury to tissue such as the skin, hyperalgesia develops. Hyperalgesia is characterized by an increase in the magnitude of pain evoked by noxious stimuli. It has been postulated that in the mechanism of hyperalgesia (especially secondary hyperalgesia) and allodynia, a sensitization of central nervous system such as spinal dorsal horn may contribute to development of hyperalgesia. However, the precise mechanism is still unclear. In the present study, we investigated the roles of N-methyl-D-aspartate (NMDA) receptor and nitric oxide (NO) system in the mechanism of hyperalgesia, and their relations with c-fos expression Methods: Inflammation was induced by injection of complete Freund adjuvant (CFA) into unilateral hindpaw of Sprague-Dawley rat. Behavioral studies measuring paw withdrawal responses by von Frey filaments and paw withdrawal latencies by radiant heat stimuli and stainings of nicotinamide adenine dinucleotide phosphate (NADPH)-diaphorase and c-fos immunoreactivity were performed. The effects of MK-801, an NMDA receptor blocker and $N^\omega$-nitro-L-arginine (L-NNA), a nitric oxide synthase (NOS) inhibitor were evaluated. Results: 1) Injection of CFA induced mechanical allodynia, mechanical hyperalgesia and thermal hyperalgesia. And it increased the number of NADPH-diaphorase positive neurons and c-fos expression neurons. 2) MK-801 inhibited mechanical hyperalgesia and thermal hyperalgesia induced by CFA and reduced the number of NADPH-diaphorase positive neurons and c-fos expression neurons. 3) L-NNA inhibited the thermal hyperalgesia and reduced the number of NADPH-diaphorase positive neurons, but did not affect the number of c-fos expression neurons. Conclusions: These results suggest that in the mechanism of mechanical hyperalgesia, NMDA receptor but not NO-system is involved and in the case of thermal hyperalgesia both NMDA receptor and NO system are involved. NO system did not affect the expression of c-fos, but c-fos expression and NOS activity were dependent on the activity of NMDA receptor.

  • PDF

Differential Role of Central GABA Receptors in Nociception of Orofacial Area in Rats

  • Lee, Ah-Ram;Lim, Nak-hyung;Kim, Hye-Jin;Kim, Min-Ji;Ju, Jin-Sook;Park, Min-Kyoung;Lee, Min-Kyung;Yang, Kui-Ye;Ahn, Dong-Kuk
    • International Journal of Oral Biology
    • /
    • v.40 no.3
    • /
    • pp.117-125
    • /
    • 2015
  • The present study investigated the role of central $GABA_A$ and $GABA_B$ receptors in orofacial pain in rats. Experiments were conducted on Sprague-Dawley rats weighing between 230 and 280 g. Intracisternal catheterization was performed for intracisternal injection, under ketamine anesthesia. Complete Freund's Adjuvant (CFA)-induced thermal hyperalgesia and inferior alveolar nerve injury-induced mechanical allodynia were employed as orofacial pain models. Intracisternal administration of bicuculline, a $GABA_A$ receptor antagonist, produced mechanical allodynia in naive rats, but not thermal hyperalgesia. However, CGP35348, a $GABA_B$ receptor antagonist, did not show any pain behavior in naive rats. Intracisternal administration of muscimol, a $GABA_A$ receptor agonist, attenuated the thermal hyperalgesia and mechanical allodynia in rats with CFA treatment and inferior alveolar nerve injury, respectively. On the contrary, intracisternal administration of bicuculline also attenuated the mechanical allodynia in rats with inferior alveolar nerve injury. Intracisternal administration of baclofen, a $GABA_B$ receptor agonist, attenuated the thermal hyperalgesia and mechanical allodynia in rats with CFA treatment and inferior alveolar nerve injury, respectively. In contrast to $GABA_A$ receptor antagonist, intracisternal administration of CGP35348 did not affect either the thermal hyperalgesia or mechanical allodynia. Our current findings suggest that the $GABA_A$ receptor, but not the $GABA_B$ receptor, participates in pain processing under normal conditions. Intracisternal administration of $GABA_A$ receptor antagonist, but not $GABA_B$ receptor antagonist, produces paradoxical antinociception under pain conditions. These results suggest that central GABA has differential roles in the processing of orofacial pain, and the blockade of $GABA_A$ receptor provides new therapeutic targets for the treatment of chronic pain.

Attenuated Neuropathic Pain in CaV3.1 Null Mice

  • Na, Heung Sik;Choi, Soonwook;Kim, Junesun;Park, Joonoh;Shin, Hee-Sup
    • Molecules and Cells
    • /
    • v.25 no.2
    • /
    • pp.242-246
    • /
    • 2008
  • To assess the role of $\alpha_{1G}$ T-type $Ca^{2+}$ channels in neuropathic pain after L5 spinal nerve ligation, we examined behavioral pain susceptibility in mice lacking $Ca_{V}3.1$ (${\alpha}_{1G}{^{-/-}}$), the gene encoding the pore-forming units of these channels. Reduced spontaneous pain responses and an increased threshold for paw withdrawal in response to mechanical stimulation were observed in these mice. The ${{\alpha}_{1G}}^{-/-}$ mice also showed attenuated thermal hyperalgesia in response to both low-(IR30) and high-intensity (IR60) infrared stimulation. Our results reveal the importance of ${\alpha}_{1G}$ T-type $Ca^{2+}$ channels in the development of neuropathic pain, and suggest that selective modulation of ${\alpha}_{1G}$ subtype channels may provide a novel approach to the treatment of allodynia and hyperalgesia.

Effects of Bupivacaine Pretreatment at the Periphery on the Expression of Mechanical Hyperalgesia in a Rat Model of Peripheral Neuropathy (백서의 말초신경 통증 모델에서 신경결찰 전 말초에 주입된 Bupivacaine이 기계적 통각과민의 발현에 미치는 영향)

  • Chung, Yong-Bo;Leem, Joong-Woo;Chung, Eun-Jung;Lee, Jung-Chan;Choi, Yoon
    • The Korean Journal of Pain
    • /
    • v.14 no.1
    • /
    • pp.7-11
    • /
    • 2001
  • Background: Although several mechanisms of causalgia, which results from a partial injury to the peripheral nerve trunk, have been proposed, whether or not antidromic impulses from the injured neurons contribute to the development of the mechanical hyperalgesia has not been studied. The purpose of this experiment is was to investigate the role of antidromic impulses to the peripheral sensory receptor site on the development of mechanical hyperalgesia in a rat model of peripheral neuropathy. Methods: Rats were prepared with tight ligation of by tightly ligating the left fifth and sixth lumbar spinal nerves. The effect of bupivacaine pretreatment on the development of mechanical hyperalgesia was evaluated by injecting 0.5% bupivacaine (0.3 ml) into the plantar surface of the left hind paw before the skin incision was made. For the control group, normal saline (0.3 ml) was injected instead of bupivacaine. To measure the mechanical hyperalgesia, paw withdrawal thresholds were measured using a series of von Frey hairs. Mechanical hyperalgesia was measured a the day before, and 1, 2, 3, 4, 7, and 14 days after the surgery. Results: The control group showed decreased withdrawal thresholds from the day after the surgery (the values were $14.0{\pm}0.5$, $8.9{\pm}1.3$, $8.4{\pm}1.6$, $6.9{\pm}1.2$, $8.8{\pm}1.5$, $10.5{\pm}1.3$, and $8.6{\pm}1.3$ g; at -1, 1, 2, 3, 4, 7, and 14 days after the surgery, respectively). However, withdrawal thresholds of the bupivacaine-pretreated group showed increased withdrawal thresholds for three days after the surgery ($14.5{\pm}0.3$, $12.6{\pm}1.4$, $12.7{\pm}1.1$, $10.5{\pm}1.3$ g; at 1, 1, 2, 3 days after the surgery). Conclusions: Our result suggests that antidromic impulses to the peripheral sensory receptors are at least partly responsible for the initial development of mechanical hyperalgesia in a rat model of peripheral neuropathy.

  • PDF

Spinal orexin A attenuates opioid-induced mechanical hypersensitivity in the rat

  • Youn, Dong-ho;Jun, Jiyeon;Kim, Tae Wan;Park, Kibeom
    • The Korean Journal of Pain
    • /
    • v.35 no.4
    • /
    • pp.433-439
    • /
    • 2022
  • Background: Repeated administration of opioid analgesics for pain treatment can produce paradoxical hyperalgesia via peripheral and/or central mechanisms. Thus, this study investigated whether spinally (centrally) administered orexin A attenuates opioid-induced hyperalgesia (OIH). Methods: [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO), a selective µ-opioid receptor agonist, was used to induce mechanical hypersensitivity and was administered intradermally (4 times, 1-hour intervals) on the rat hind paw dorsum. To determine whether post- or pretreatments with spinal orexin A, dynorphin A, and anti-dynorphin A were effective in OIH, the drugs were injected through an intrathecal catheter whose tip was positioned dorsally at the L3 segment of the spinal cord (5 ㎍ for all). Mechanical hypersensitivity was assessed using von Frey monofilaments. Results: Repeated intradermal injections of DAMGO resulted in mechanical hypersensitivity in rats, lasting more than 8 days. Although the first intrathecal treatment of orexin A on the 6th day after DAMGO exposure did not show any significant effect on the mechanical threshold, the second (on the 8th day) significantly attenuated the DAMGO-induced mechanical hypersensitivity, which disappeared when the type 1 orexin receptor (OX1R) was blocked. However, intrathecal administration of dynorphin or an anti-dynorphin antibody (dynorphin antagonists) had no effect on DAMGO-induced hypersensitivity. Lastly, pretreatment with orexin A, dynorphin, or anti-dynorphin did not prevent DAMGO-induced mechanical hypersensitivity. Conclusions: Spinal orexin A attenuates mechanical hyperalgesia induced by repetitive intradermal injections of DAMGO through OX1R. These data suggest that OIH can be potentially treated by activating the orexin A-OX1R pathway in the spinal dorsal horn.