用户名: 密码: 验证码:
NR2B在小鼠骨癌痛中的作用及机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的建立新的小鼠骨癌痛模型,观察NMDA受体2B亚基(NR2B)在骨癌痛小鼠前脑扣带回和脊髓中的表达及分布情况;明确NR2B与癌痛产生之间的关系;探讨NR2B在小鼠骨癌痛中的作用机制。
     方法
     1.小鼠骨癌痛新模型的制备及评价:选用成年雄性C57BL/6小鼠30只,随机分为4组,模型组(15只):接种Lewis肺癌细胞2×106个;热灭活组(5只):接种相同数目加热灭活的Lewis肺癌细胞;PBS组(5只):接种等容积的PBS液;正常对照组(5只):不予任何处理。接种后7天始隔日观察小鼠自发痛行为及测定机械性触诱发痛。第7、15、23天,将小鼠麻醉后行双侧后肢X线摄片,评估肿瘤诱发的骨组织破坏程度。模型组小鼠每次摄片后任选5只取术侧后肢,各对照组小鼠最后一次摄片后取术侧后肢,分别作苏木素-伊红(HE)染色观察骨质破坏情况。
     2.NR2B在骨癌痛小鼠前扣带回和脊髓中的分布与表达:选用成年雄性C57BL/6小鼠81只,随机分为3组:癌痛组,假手术组和正常对照组。观察三组小鼠自发痛行为及测定机械性触诱发痛。分别于术后第6、10、14、18、22天各取3只,实时定量PCR检测前扣带回和脊髓NR2B mRNA的表达。依据PCR结果,每组分别于术后第14、18天各取3只,免疫组化检测前扣带回和脊髓NR2B的分布和表达;同时每组另各取3只,于术后第14、18天行Western Blot检测脊髓NR2B蛋白的表达。
     3.脊髓NR2B基因沉默对小鼠骨癌痛的影响:选用成年雄性C57BL/6小鼠72只,随机分为3组:癌痛组,干扰组和干扰对照组。观察各组小鼠自发痛行为及测定机械性触诱发痛。每组分别于接种后第13、14、15、16、18、22天各取3只,实时定量PCR检测脊髓NR2B mRNA的表达。依据PCR的结果,于接种后第14和18天每组各取3只,Western Blot检测脊髓NR2B蛋白的表达。
     4.MAPK信号通路介导的NR2B在小鼠骨癌痛中的作用机制:选用成年雄性C57BL/6小鼠48只,随机分为4组:正常组,癌痛组,干扰组和干扰对照组。观察各组小鼠自发痛行为及测定机械性触诱发痛。每组分别于接种后第14和18天各取3只,实时定量PCR检测脊髓NR2B、SP、c-fos和GFAP基因表达水平。选取接种后第14和18天的每组小鼠各3只,Western Blot检测脊髓NR2B、p-ERK、p-p38和GFAP蛋白表达水平。
     结果
     1.小鼠骨癌痛新模型的评价:模型组接种后第11天左右出现明显自发痛行为,表现为自发抬足时间延长;接种后第13天左右出现明显的触诱发痛,表现为50%缩足阈值明显下降,且持续整个观察期。接种后第23天放射学结果显示,术侧股骨下段骨髓腔消失,骨皮质中断、大块缺损。同时组织学可见肿瘤细胞充满骨髓腔,且穿破骨皮质向外生长,侵犯周围肌肉组织。行为学、放射学、组织学三方面证实建模成功。
     2.NR2B在骨癌痛小鼠的前扣带回和脊髓中的分布与表达:
     (1)前扣带回NR2B的实时荧光定量PCR结果显示:接种后第6天、10天,癌痛组NR2B的表达与假手术组比较无显著差异,而癌痛组和假手术组NR2B的表达比正常组明显增高;第14天、18天,癌痛组NR2B的表达明显增加,与假手术组和正常组比较,有显著差异(p<0.05),而假手术组与正常组比较,无显著差异(p>0.05);第22天,癌痛组NR2B的表达下降,与假手术组和正常组比较无统计学意义。L4-6段脊髓NR2B的实时荧光定量PCR示:接种后第6天,癌痛组NR2B的表达与假手术组比较无显著差异,而癌痛组和假手术组NR2B的表达比正常组明显增高;第10、14、18、22天,癌痛组NR2B的表达明显增加,与假手术组和正常组比较,有显著差异(p<0.05);而假手术组与正常组比较,无显著差异(p>0.05)。
     (2)L4-6段脊髓和前扣带回NR2B免疫组化测定结果:癌痛组NR2B主要分布在脊髓背角浅层、中央导水管、前角以及小鼠的前扣带回,脊髓各层NR2B表达量在中央导水管、背角浅层、前角依次增多。癌痛组于术后第14、18天,脊髓和手术对侧的前扣带回区NR2B阳性细胞光密度(OD)值与假手术组和正常组比较有统计学差异(P<0.05);而假手术组与正常组比较,无显著差异(p>0.05)。
     (3)L4-6段脊髓NR2B表达的Western Blot检测结果:癌痛组于接种后第14天、18天,脊髓NR2B的表达明显增高,与假手术组和正常组比较,有显著差异(p<0.05);而假手术组与正常组比较,无显著差异(p>0.05)。
     3.脊髓NR2B基因沉默对小鼠骨癌痛的影响:
     (1)实时定量PCR检测小鼠脊髓NR2B mRNA的表达:接种后第14天和第18天干扰组小鼠L4-6段脊髓的NR2B mRNA表达水平比癌痛组明显降低,其中第14天下调最明显,干扰组比癌痛组NR2B的表达下调了74%;而干扰对照组与癌痛组比较,无显著差异(p>0.05)。
     (2)Western Blot检测小鼠脊髓NR2B蛋白的表达:接种后第14和18天干扰组小鼠L4-6段脊髓的NR2B表达水平比癌痛组明显降低,其中第18天下调最明显,干扰组比癌痛组NR2B表达下调了67%;而干扰对照组与癌痛组比较,无显著差异(p>0.05)。
     (3)干扰组与其他两组相比,自发抬足时间明显缩短,触诱发痛阈值明显增加,有显著差异(p<0.05)。
     4.MAPK信号通路介导的NR2B在小鼠骨癌痛中的作用机制:
     (1)实时定量PCR检测各组小鼠L4-6段脊髓的NR2B、SP、c-fos和GFAP基因mRNA的表达:接种术后第14、18天癌痛组小鼠脊髓的NR2B、SP、c-fos和GFAP基因表达比正常组明显上调(p<0.05);干扰组NR2B、SP、c-fos和GFAP基因表达比癌痛组显著降低(p<0.05);而干扰对照组与癌痛组比较,无显著差异(p>0.05)。
     (2)Western Blot检测各组小鼠L4-6段脊髓的NR2B、p-ERK、p-p38和GFAP蛋白的表达:接种后第14、18天癌痛组小鼠脊髓的NR2B、p-ERK、p-p38和GFAP的蛋白表达比正常组明显上调(p<0.05);干扰组NR2B、p-ERK、p-p38和GFAP的蛋白表达比癌痛组显著降低(p<0.05);而干扰对照组与癌痛组比较,无显著差异(p>0.05)。
     结论:
     1.C57BL/6小鼠股骨骨髓腔接种Lewis肺癌细胞能成功制备小鼠骨癌痛新模型,此模型与人类骨癌痛具有相似性;
     2.NR2B在小鼠骨癌痛模型中呈高表达,且同一癌痛模型中观察到NR2B在L4-L6段脊髓和前扣带回同时高表达,提示在癌痛的产生和维持中不仅激活了脊髓低位中枢,而且有脑部中枢参与;
     3.本研究初次发现NR2B在癌痛模型的L4-6段脊髓前角细胞表达呈强阳性,为下一步研究提供了新的思路;
     4.PEI/NR2B-siRNA复合物能特异性的使骨癌痛小鼠脊髓NR2B的表达明显下调;其表达下调与小鼠疼痛行为学减轻相一致,进一步证明NR2B在骨癌痛发生过程中起重要作用;
     5.MAPK信号转导通路(ERK通路和p38通路)在骨癌痛小鼠模型中被激活;NR2B参与了MAPK信号通路介导的骨癌痛的中枢敏化;
     6.NR2B参与介导的骨癌痛可能与GFAP的过表达相关,其机制有待进一步研究。
Objective:In order to investigate the mechanisms of bone cancer pain, a mouse bone cancer pain model is established to study the role of NR2B in the initiation and maintenance of bone cancer pain by observing the expression and distribution of NR2B in the anterior cingulate cortex and L4-6 spinal cord of the animal.
     1. The establishment and evaluation of the new model of mouse cancer pain:thirty adult male C57BL/6 mice were randomly assigned to four groups:the test group (n=15):10μl PBS containing 2×106 Lewis lung cancer cells were inoculated into the marrow cavity of distal femur of the animal; the heat-inactivated group (n=5):the same number of Lewis lung cancer cells that had been heat-inactivated were inoculated; PBS group (n=5):the same volume of PBS solution was inoculated; normal control group (n=5):no treatment. We observed the spontaneous lifting time and mechanical allodynia of mice on alternate days from the 7th day after inoculation. The structural damage of the operated femur induced by tumor was monitored by radiological analysis by comparing with the other femur bone of the animal and observed respectively on the 7th,15th,23rd day. In the test group, five femurs from operated animals that were randomly selected were stained with hematoxylin and eosin (HE) after radiography; bone destruction was observed on 7th,15th,23rd day, respectively. Other groups were observed only on the 23rd day.
     2. The expression and distribution of NR2B in the anterior cingulate cortex and spinal cord in the test group:eighty-one adult male C57BL/6 mice were randomly assigned to three groups:cancer pain group (N=27); sham-operated group (N=27); normal control group (N=27).10μl of D-Hanks solution containing 2×106 Lewis lung cancer cells were inoculated in the cancer pain group; the same volume of solution without cancer cells was used in the sham-operated group; the normal control group:no treatment. Spontaneous lifting time and mechanical allodynia of mouse hind paw were measured on the 6th,10th,14th,18th,22nd day after inoculation. Three mice were taken to observe the expression of NR2B mRNA by using the qRT-PCR detection at the same time. According to the results of the PCR, three mice from each group were decapitated to extract the L4-6 spinal cord and anterior cingulate cortex to do fluorescence quantitative RT-PCR and immunohistochemical staining to observe the expression and distribution of NR2B, the remaining three mice were used to detect NR2B protein by Western Blot on the 14th and 18th day after inoculation.
     3. The effects of NR2B gene silencing in bone cancer pain mouse: 72 adult male C57BL/6 mice were randomly divided into three groups: cancer pain group (N=21):Lewis lung cancer cells 2×106/10ul were inoculated as stated above; interference group (N=21):PEI/NR2B siRNA complex was intrathecally injected 12 days later after cancer cell inoculation; control group of the interference (N=21):the PEI/Negative control siRNA complex was used in intrathecal injection. Spontaneous lifting time and mechanical allodynia were measured pre-operation and on the 6th,10th,12th,13th,14th,15th,16th,18th and 22nd day after inoculation. L4-6 spinal cord segments were isolated for fluorescence quantitative RT-PCR and Western Blot to detect the expression levels of NR2B in each group.
     4. The effects of NR2B were mediated by MAPK signaling pathway in the mouse bone cancer pain:forty-eight adult male C57BL/6 mice were randomly divided into four groups:normal group, cancer pain group, interference group and control of interference group. Spontaneous lifting time and mechanical allodynia in each group were measured pre-inoculation and on the 10th,12th,14th,16th and 18th day after inoculation. L4-6 spinal cord of the animals from each group was used to detect the expression levels of NR2B, SP, c-fos and GFAP gene by real-time quantitative PCR, and the expression levels of NR2B, p-ERK, p-p38 and GFAP protein were detected by Western Blot on the 14th and 18th day.
     1. Evaluation of bone cancer pain model:compared with the controls, the test group displayed a gradual development of spontaneous pain showing longer flinch time from the 11th day on; there was obvious mechanical allodynia from the 13th day on after inoculation as demonstrated by a 50% reduction of von Frey threshold. On the 23rd day after operation, X-ray showed that medullary cavity of lower end of femur on ipsilateral side had disappeared and some cortical bone had been lost. Pathological examination showed that tumor cells penetrated the medullary canal and invaded peripheral tissues. Therefore, the model was validated by data from the above three aspects.
     2. The expression and distribution of NR2B in the anterior cingulate cortex and spinal cord of mouse with bone cancer pain:
     Real-time quantitative florescence PCR detection in the anterior cingulate cortex:there was no significant difference between the cancer pain group and the sham operated group on the 6th and 10th day after inoculation, but the NR2B expression in both groups was higher than that in the control group; on the 14th and 18th day, the expression of NR2B in the cancer pain group increased significantly as compared to the groups of normal control and the sham operated (p<0.05), while there was no significant difference between the sham operated group and the normal control group. On the 22nd day, the expression of NR2B in the cancer pain group was reduced, but there was no statistical significance as compared to the other two groups. Real-time quantitative florescence PCR detection of NR2B expression in L4-6 spinal cord:on the 6th day after inoculation, there was no significant difference between the cancer pain group and the sham operated group, but the expression of NR2B in these two groups was increased significantly as compared to the normal control group. On the 10th,14th,18th and 22nd day, the expression of NR2B in the cancer pain group was increased significantly as compared to the groups of normal control and the sham operated (p<0.05), while there was no significant difference between the sham operated and the normal control group.
     (2) The results of immunohistochemical examination in the anterior cingulate cortex and L4-6 spinal cord:the expression of NR2B in the cancer pain group was mainly located in the superficial dorsal horn, central aqueduct, anterior horn of the spinal cord and the anterior cingulate cortex, the expression level of NR2B was highest in the anterior horn, followed bysuperficial dorsal horn and lowest levels were observed in the central aqueduct. The optical density (OD) values of NR2B-positive cells in L4-6 spinal cord and area of contralateral anterior cingulate cortex in the cancer pain group were significantly different as compared to the sham-operated and the normal controls on the 14th and 18th day after inoculation (P<0.05), whereas there was no significant difference between the later two groups (P>0.05).
     (3) The results of the Western Blot in L4-6 spinal cord:the expression of NR2B in the spinal cord of cancer pain group was significantly increased as compared to the groups of the normal control and the sham operated on day 14 and 18 after inoculation (p<0.05), while there was no significant difference between the later two groups (P>0.05).
     3. The effects of NR2B gene silencing in cancer bone pain mouse:
     (1) Real-time quantitative PCR detection:the expression of NR2B in L4-6 spinal cord in the interference group was reduced significantly as compared to the cancer pain group and the control group of the interference group on the 14th and 18th day after inoculation (P<0.05). The most obvious change in the expression of NR2B in the interference group was observed on the 14th day after inoculation, showing 74% lower expression in the test group as compared to that in the cancer pain group and the control group of the interference. There was no significant difference between the later two groups (P>0.05).
     (2) The results of Western Blot:the expression of NR2B in the L4-6 spinal cord in the interference group was reduced significantly as compared to that in the cancer pain group on the 14th and 18th day after inoculation (P<0.05). The most obvious change in the expression of NR2B was observed on the 18th day after inoculation, demonstrating 67% lower expression in the interference group than that in the cancer pain group. There was no significant difference between the later two groups (P>0.05).
     (3) Spontaneous lifting time was decreased and mechanical allodynia was increased obviously in the interference group as compared to the cancer pain group and the control group of the interference.
     4. The effects of NR2B were mediated by MAPK signaling pathway in bone cancer pain in mouse:
     (1) Real-time quantitative PCR detection:the expression levels of NR2B, SP, c-fos and GFAP genes in the L4-6 spinal cord of cancer pain group was significantly increased as compared to the normal control group on the 14th and 18th day after inoculation (P<0.05); the expression of these genes in the L4-6 spinal cord in the interference group was significantly lower than that of the cancer pain group; while there was no significant difference between the cancer pain group and the control of interference group(p>0.05).
     (2) Western Blot detection results:the expression of NR2B, p-ERK, p-p38 and GFAP protein in the spinal cord of cancer pain group was significantly increased as compared to that in the normal group on the 14th and 18th day after inoculation (P<0.05); the expression of these proteins in the L4-6 spinal cord of the interference group was significantly lower than that in the cancer pain group (P<0.05); while there was no significant difference between the cancer pain group and the control group of interference group(p>0.05).
     1. Inoculation of Lewis lung cancer cells into the marrow cavity of distal femur of C57BL/6 mice can effectively establish a mouse bone cancer pain model. There was much similarity between human cancer pain and this model.
     2. The observation that NR2B was expressed in large quantity in both the L4-6 spinal cord and anterior cingulate cortex suggests that the generation and maintenance of bone cancer pain not only can be activated in the spinal cord, but also is related to the anterior cingulate cortex.
     3. In this study we found that the expression of NR2B was strongly positive in the anterior horn cells of spinal cord of the bone cancer pain model, which provided some new insights about the mechanism of cancer pain and might be useful in later research.
     4. The expression of NR2B was reduced in the L4-6 spinal cord after intrathecal injection of PEI/NR2B-siRNA complex in the mouse with cancer pain, which was consistent with the behavior changes of the animals, and provided further evidence showing that NR2B might play an important role in the initiation of bone cancer pain.
     5. The MAPK signaling pathway was activated in the bone cancer pain model, suggesting that MAPK signal transduction pathway (ERK pathway and p38 pathway) is involved in the generation and maintenance of bone cancer pain.
     6. Bone cancer pain is mediated by NR2B receptor system, and may be related to the over-expression of GFAP, yet the mechanisms involved need to be further explored.
引文
[1]Banning A, Sjogren P, Henriksen H. Pain causes in 200 patients referred to a multidisciplinary cancer pain clinic. Pain,1991,45:45-48.
    [2]Rubens, RD,. Bone metastases—the clinical problem. Eur J Cancer,1998,34: 210-213.
    [3]Mantyh PW, Clohisy DR, Koltzenburg M, Hunt SP. Molecular mechanisms of cancer pain. Nat Rev Cancer,2002,2:201-209.
    [4]Meuser T, Pietruck C, Radbruch L, et al. Symptoms duringcancer pain treatment following WHO-guidelines:a longitudinal follow-up study of symptom prevalence, severity and etiology. Pain,2001,93:247-257.
    [5]Schwei MJ, Honore P, Rogers SD, et a. Neurochemical and cellular reorganization of the spinal cord in a murine model of bone cancer pain. J Neurosci,1999,19: 10886-10897.
    [6]Kawasaki Y, Kohno T, Zhuang ZY, et al. Ionotropic and metabotropic receptors, protein kinase A, protein kinase C, and Src contribute to C-fiber-induced ERK activation and cAMP response element-binding protein phosphorylation in dorsal horn neurons, leading to central sensitization. J Neurosci,2004,24:8310-8321.
    [7]Khasabov SG, Rogers SD, Ghilardi JR, et al. Spinal neurons that possess the substance P receptor are required for the development of central sensitization. J Neurosci,2002,22:9086-9098.
    [8]Andrei B. Petrenko, Tomohiro Yamakura, Hiroshi Baba, et al. The Role of N-Methyl-D-Aspartate (NMDA) Receptors in Pain:A Review. Anesth Analg, 2003,97:1108-1116.
    [9]Petrenko, Andrei B. Yamakura, et al. The role of N-Methyl-d-Aspartate (NMDA) receptors in pain. Anesth Analg.2003,97(4):1108-1116.
    [10]Kew JN, Kemp JA. Ionotropic and metabotropic glutamate receptor structure and pharmacology. Psychopharmacology (Berl),2005,179:4-29.
    [11]Nagy GG, Watanabe M, Fukaya M, et al. Synaptic distribution of the NR1, NR2A and NR2B subunits of the N-methyl-D-aspartate receptor in the rat lumbar spinal cord revealed with an antigen-unmasking techenique. Eur J Neurosci, 2004,20 (12):3301-3312.
    [12]Ma QP, Hargreaves RJ. Localization of N-methyl-D-aspartate NR2B subunison primary sensory neurons that give rise to small-caliber sciatic nerve fibers in rats. Neuroscience,2000,101 (3):699-707.
    [13]Boyce S, Wyatt A, Webb, JK, et al. Selective NMDA NR2B antagonists induce antinociception without motor dysfunction correlation with restricted localistation of NR2B subunit in dorsal horn. Neuropharmacology,1999,38 (5): 611-623.
    [14]Petrenko AB, Yamakura T, Baba H, et al. Unaltered pain-related behavior in mice lacking NMDA receptor GluR epsilon 1 subunit. Neurosci Research,2003, 46:199-204.
    [15]Tan PH, Yang LC, Shin HC, et al. Gene knockdown with intrathecal siRNA of NMDA receptor NR2B subunit reduces formalin-induced nociception in the rat. Gene Therapy,2005,12(1):59-66.
    [16]De Leo JA, Tawfik VL, LaCroix-Fralish ML. The tetrapartite synapse:path to CNS sensitization and chronic pain. Pain,2006,122 (122):17221.
    [17]Moalem G, Tracey DJ. Immune and inflammatorymechanisms inneuropathic pain. Brain Res Rev,2006,51(2):240-264.
    [18]Honore P, Rogers SD, Schwei MJ, Salak-Johnson JL, Luger NM, Sabino MC, Clohisy DR, Mantyh PW. Murine models of inflammatory, neuropathic and cancer pain each generates a unique set of neurochemical changes in the spinal cord and sensory neurons. Neuroscience,2000,98:585-598.
    [19]Kjonniksen I, Winderen M, Brutand O, et al. Validity and usefulness of human tumor models established by intratibial cell inoculation in nude rats. Cancer Res, 1994,54:1715-1719.
    [20]Medhurst SJ, Walker K, Bowes M, et al. A rat model of bone cancer pain. Pain, 2002,96 (1-2):129-140
    [21]Mao-Ying Q, Zhao J, Dong ZQ, et al. A rat model of bone cancer pain induced by intra-tibia inoculation of Walker 256 mammary gland carcinoma cells. Biochem Biophys Res Commun,2006,14,345(4):1292-1298.
    [22]董航,田玉科,项红兵.大鼠胫骨癌痛模型的制备及评价.中国临床康复,2006,10(20):77-79.
    [23]Taskinen HS, Roytta M. Increased expression of chemokines (MCP-1, MIP-1 alpha, RANTES) after peripheral nerve transaction. J Peripher Nerv Syst, 2000,5(2):75-81.
    [24]Vicent S, Luis-Ravelo D, Anton, et al. A novel lung cancer signature mediates
    metastatic bone colonization by a dual mechanism.Cancer Research, 2008,68(7):2275-2285.
    [25]Chaplan SR, Bach FW, Pogrel JW et al. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods,1994,53:55-63.
    [26]Dixon WJ. Efficient analysis of experimental observations. Annu. Rev. Pharmacol. Toxicol,1980,20:441-462.
    [27]Honore P, Schwei J, Rogers SD, et al. Cellular and neurochemical remodeling of the spinal cord in bone cancer pain. Prog Brain Res,2000,129:389-397.
    [28]Martin TJ, Eisenach JC. Pharmacology of opioid and nonopioid analgesics in chronic pain states. J Pharmacol Exp Ther,2001,299:811-817.
    [29]Mouedden EL, Meert TF. Evaluation of pain-related behavior, bone destruction and effectiveness of fentanyl, sufentanil, and morphine in a murine model of cancer pain. Pharmacol Biochem Behav,2005,82:109-119.
    [30]Luger NM, Sabino MA, Schwei MJ, et al. Efficacy of systemic morphine suggests a fundamental difference in the mechanisms that generate bone cancer vs inflammatory pain. Pain,2002,99:397-406.
    [31]Mohammed El Mouedden, Theo Frans Meert. Pharmacological evaluation of opioid and non-opioid analgesics in a murine bone cancer model of pain. Pharmacol Biochem and Behav,2007,86(3):458-467.
    [32]Petersa CM, Ghilardid JR, Keysera CP, et al. Tumor-induced injury of primary afferent sensory nerve fibers in bone cancer pain. Exp Neurol,2005,193(1): 85-100.
    [33]Sasamura T, Nakamura S, Lida Y, et al. Morphine analgesia suppresses tumor growth and metastasis in a mouse model of cancer pain produced by orthotopic tumor inoculation. Eur J Pharmacol.2002,441(3):185-191.
    [34]Kehl LJ, Hamamoto DT, Wacnik PW, et al. A cannabinoid agonist differentially attenuates deep tissue hyperalgesia in animal models of cancer and inflammatory muscle pain. Pain,2003,103(1-2):175-186.
    [35]Jennifer M, Loftis, Aaron Janowsky. The N-methyl-D-aspartate receptorsubunit NR2B:localization, functional properties, regulation, and clinical implications. Pharmacology &Therapeutics,2003,97:55-85.
    [36]Minoru Narita, Kan Miyoshi, Michiko Narita, et al. Changes in function of NMDA receptor NR2B subunit in spinal cord of rats with neuropathy following chronic ethanol consumption. Life Sciences,2007,80:852-859.
    [37]Liu H, Wang H, Sheng M, et al. Evidence for presynaptic N-methyl-D-aspartate autoreceptors in the spinal cord dorsal horn. PNAS,1991,91(5):9383-9387.
    [38]Hideaki Iwatal, Masao Maekawa, Yuuichi Hori. Differential expression of NMDA receptor subunits between neurons containing and not containing enkephalin in the mouse embryo spinal cord. Neuroscience Letters,2005, 391(1-2):11-16.
    [39]Luque JM, Bleuel Z, Malherbe P, et al. Alternatively spliced isoforms of the N-methyl-D-aspartate receptor subunit are differentially distributed within the rat spinal cord. Neuroscience,1994,63(90):629-635.
    [40]Woolf CJ. Evidence for a central component of post-injury pain hypersensitivity. Nature,1983,306 (5944):686-688.
    [41]Portenoy RK, Lesage P. Management of cancer pain. Lancet,1999,353: 1695-1700.
    [42]Etsuko Nakazato, Aki Kato, Shuzo Watanabe Brain but Not Spinal NR2B Receptor Is Responsible for the Anti-Allodynic Effect of an NR2B Subunit-Selective Antagonist CP-101,606 in a Rat Chronic Constriction Injury Model. Pharmacology,2005,73:8-14.
    [43]Wei F, Wang GD Kerchner GA, et al. Genetic enhancement of in flammatory pain by forebrain NR2B overexpressiont. Nature neuroscience,2001,4(1):164-169.
    [44]Apkarian AV, Bushnell MC, Treede RD, et al. Human brain mechanisms of pain perception and regulation in health and disease. Eur J Pain,2005,9:463-484.
    [45]Mohr C, Binkofskia F, Erdmann C, et al. The anterior cingulate cortex contain distinct areas dissociating external from self-administered painful stimulation:a parametric fMRI study. Pain,2005,114:347-357.
    [46]Buchel, Raymond, Dolan J, Jorge L, et al. Amygdala-Hippocampal Involvement in Human Aversive Trace Conditioning Revealed through Event-Related Functional Magnetic Resonance Imaging. Journal of Neuroscience,1999,19(24): 10869-10876.
    [47]Fan J, Wu X, Cao Z, et al. Up-regulation of anterior cingulate cortex NR2B receptors contributes to visceral pain responses in rats. Gastroenterology,2009, 136(5):1732-1740.
    [48]Meuser T, Pietruck C, Radbruch L, et al. Symptoms during cancer pain treatment following WHO-guidelines:a longitudinal follow-up study of symptom
    prevalence, severity and etiology. Pain,2001,93:247-257.
    [49]Fire A, Xu S, Montgomery MK, et al. Potent andspecific genetic interference by double-stranded RNA in Caenorhabdits elegans. Nature,1998,391:806-811.
    [50]Zamore PD, Tuschl T, Sharp PA, et al. RNAi:double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell, 2000,101:25-33.
    [51]Elbashir SM, Harborth J, Weber K, Tuschl T. Analysis of gene function in somatic ammalian cells using small interfering RNAs. Methods,2002,26: 199-213.
    [52]Elbashir SM, Lendeckel W, Tuschl T. RNA interference is mediated by 21-and 22-nucleotide RNAs. Genes & Development,2001,15 (2):188-200.
    [53]Elbashir SM, Martinez J, Patkaniowska A, et al. Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate. EMBO J,2001,20:6877-6888.
    [54]Goblirsch M, Mathews W, Lynch C, et al. Radiation treatment decreases bone cancer pain, osteolysis and tumor size. Radiat Res,2004.161:228-234.
    [55]Schwei MJ, Honore P, Rogers SD, et al. Neurochemical and cellular reorganization of t he spinal cord in a murine model of bone cancer pain. J Neurosci,1999,19:10886-10897.
    [56]Lehre KP, Danbolt NC. The number of glutamate t ransporter subtype molecules at glutamatergic synapses:chemical and stereological quantification in young adult rat brain. J Neurosci,1998,18:8751-8757.
    [57]Watkins LR, Maier SF. Glia:a novel drug discovery target for clinical pain. Nat Rev Drug Discov,2003,2:973-985.
    [58]Honore P, Luger NM, Sabino MA, et al. Osteoprotegerin blocks bone cancerinduced skeletal destruction, skeletal pain and painrelated neurochemical reorganization of the spinal cord. Nat Med,2000,6:521-528.
    [59]Goblirsch M, Mathews W, Lynch C, et al. Radiation treatment decreases bone cancer pain, osteolysis and tumor size. Radiat Res,2004,161:228-234.
    [60]Svensson CI, Fitzsimmons B, Azizi S, et al. Spinal p38beta isoform mediates tissue injury induced hyperalgesia and spinal sensitization. J Neurochem,2005, 92(6):1508-1520.
    [1]Portenoy RK, Lesage P. Management of cancer pain. Lancet,1999,353: 1695-1700.
    [2]Coleman RE. Skeletal complications of malignancy. Cancer,1997,80:1588-1594.
    [3]Clohisy DR, Mantyh PW. Bone cancer pain. Cancer,2003,97:866-873.
    [4]S. Mercadante, Malignant bone pain:pathophysiology and treatment,Pain,1997 69:1-18.
    [5]Kjonniksen I, Winderen M, Brutand O, et al. Validity and usefulness of human tumor models established by intratibial cell inoculation in nude rats. Cancer Res, 1994,54:1715-1719.
    [6]Schwei MJ, Honore P, Rogers SD, et al. Neurochemical and cellular reorganization of the spinal cord in a murine model of bone cancer pain. J Neurosci,1999,19:10886-10897.
    [7]Medhurst SJ, Walker K, Bowes M, et al. A rat model of bone cancer pain. Pain, 2002,96:129-140.
    [8]Wacnik PW, Eikmeier LJ, Ruggles TR, et al. Functional interactions between tumor and peripheral nerve:morphology, algogen identification, and behavioral characterization of a new murine model of cancer pain. Neurosci, 2001,21:9355-9366.
    [9]Sarlak A, Gundes H, Ozkurkcugil C, et al. Solitary calcanealmetastasis in superficial bladder carcinoma. Int Clin Pract,2000,54:681-682.
    [10]Honore P, Mantyh PW. Bone cancer pain:Frommechanism to model to therapy. Pain,2000,1:303-309.
    [11]Cain DM, Wacnik PW, Eikmeier L, et al. Functional interactions between tumor and peripheral nerve in a model of cancer pain in the mouse. Pain 2001,2:15-23.
    [12]Mach DB, Rogers SD, Sabino MC, et al. Origins of skeletal pain:Sensory and sympathetic innervation of the mouse femur. Neuroscience,2002,113:155-166.
    [13]Wacnik PW, Kehl LJ, Trempe TM, et al. Tumor implantation in mouse humerus evokes movement-related hyperalgesia exceeding that evoked by intramuscular carrageenan. Pain,2003,101:175-186.
    [14]Goblirsch M, Mathews W, Lynch C, et al. Radiation treatment decreases bone cancer pain, osteolysis and tumor size. Radiat Res,2004,161:228-234.
    [15]Hukkanen M, Konttinen YT, Rees RG, et al. Distribution of nerve endings and sensory neuropeptides in rat synovium, meniscus and bone. Int J Tissue React,
    1992,14:1-10.
    [16]Sevcik MA, Luger NM, Mach DB, et al. Bone cancer pain:The effects of the bisphosphonate alendronate on pain, skeletal remodeling, tumor growth and tumor necrosis. Pain,2004,111:169-180.
    [17]Urch C. The pathophysiology of cancer-induced bone pain:Current understanding. Palliat Med,2004,18:267-274.
    [18]Galeazza MT, Garry MG, Yost HJ, et al. Plasticity in the synthesis and storage of substance P and calcitonin gene-related peptide in primary afferent neurons during peripheral inflammation. Neuroscience,1995,66:443-458.
    [19]Garrison CJ, Dougherty PM, Carlton SM. Quantitative analysis of substance P and calcitonin gene-related peptide immunohistochemical staining in the dorsal horn of neuropathic MK-801-treated rats. Brain Res,1993.607:205-214.
    [20]Zhang X, Bean AJ, Wiesenfeld-Hallin Z, et al. Ultrastructural studies on peptides in the dorsal horn of the rat spinal cord—Ⅲ. Effects of peripheral axotomy with special reference to galanin. Neuroscience,1995,64:893-915.
    [21]Honore P, Luger NM, Sabino MA, et al. Osteoprotegerin blocks bone cancerinduced skeletal destruction, skeletal pain and painrelated neurochemical reorganization of the spinal cord. Nat Med,2000,6:521-528.
    [22]Honore P, Rogers SD, Schwei MJ, et al. Murine models of inflammatory, neuropathic and cancer pain each generates a unique set of neurochemical changes in the spinal cord and sensory neurons. Neuroscience,2000,98: 585-598.
    [23]Djouhri L, Bleazard L, Lawson SN. Association of somatic action potential shape with sensory receptive properties in guinea-pig dorsal root ganglion neurones. J Physiol,1998,513:857-872.
    [24]Luger NM, Honore P, Sabino MAC et al. Osteoprotegerin diminishes advanced bone cancer pain. Cancer Res,2001,61:4038-4047.
    [25]Donovan, Rodriguez T, Dickenson AH, et al. Gabapentin normalizes spinal neuronal responses that correlate with behavior in a rat model of cancer-induced bone pain. Anesthesiology,2005,102(11):132-140.
    [26]Peters CM, Ghilardi JR, Keyser CP, et al. Tumor-induced injury of p rimary afferent sensory nerve fibers in bone cancer pain. Exp Neurol,2005,193(1): 85-100.
    [27]SabinoMA, LugerNM, Mach DB, et al. Different tumors in bone each give rise to a distinct pattern of skeletal destruction, bone cancer-related pain behaviors and neurochemical changes in the central nervous system. Int J Cancer,2003,104(5): 550-558.
    [28]Nagae M, Hiraga T, Yoneda T, et al. Acidic microenvironment created by osteoclasts causes bone pain associated with tumor colonization. J BoneMinerMetab,2007,25(2):99-104.
    [29]Kingsley LA, Fournier PG, Chirgwin JM, et al. Molecular biology of bone metastas. Mol Cancer Ther,2007,6(10):2609-2617.
    [30]Schmidt BL, Pickering V, Liu S, et al. Peripheral endothelin A recep tor antagonism attenuates carcinoma induced pain. Eur J Pain,2007,11(4):406-414.
    [31]Dougall WC, Chaisson M. The RANK/RANKL/OPG triad in cancer induced bone diseases. Cancer Metastasis Rev,2006,25(4):541-549.
    [32]Pederson L, Winding B, Foged NT, et al. Identification of breast cancer cell line derived paracrine factors that stimulate osteoclastactivity. Cancer Res,1999, 59(22):5849-5855.
    [33]Wacnik PW, EikmeierLJ, Simone DA, et al. Nocicep tive characteristics of tumor necrosis factor-a in naive and tumor-bearing mice. Neuroscience,2005, 132(2):479-491.
    [34]Helmlinger G, Sckell A, Dellian M et al. Acid production in glycolysis-impaired tumors provides new insights into tumor metabolism. Clin Cancer Res,2002,8: 1284-1291.
    [35]Julius D, Basbaum AI. Molecular mechanisms of nociception. Nature,2001,413: 203-210.
    [36]Tominaga M, Caterina MJ, Malmberg AB et al. The cloned capsaicin receptor integrates multiple pain-producing stimuli. Neuron,1998,21:531-543.
    [37]Sutherland S, Cook S, Ew M. Chemical mediators of pain due to tissue damage and ischemia. Prog Brain Res,2000,129:21-38.
    [38]Clohisy DR, Perkins SL, Ramnaraine ML. Review of cellular mechanisms of tumor osteolysis. Clin Orthop Res,2000,373:104-114.
    [39]Kinder M, Chislock E, Bussard KM, et al. Metastatic breast cancer induces an osteoblast inflammatory response. Exp Cell Res,2008,314(1):173-183.
    [40]Olson TH, Riedl MS, Vulchanova L, et al. An acid sensing ion channel (ASIC) localizes to small primary afferent neurons in rats. Neuroreport,1998,9: 1109-1113.
    [41]Guo A, Vulchanova L, Wang J, et al. Immunocytochemical localization of the vanilloid receptor 1 (VR1):relationship to neuropeptides, the P2X3 purinoceptor and IB4 binding sites. Eur J Neurosci,1999,11:946-958.
    [42]Roudier MP, Bain SD, Dougall WC, et al. Effects of the RANKL inhibitor, osteoprotegerin, on the pain and histopathology of bone cancer in rats. Clin Exp Metast,2006,23(324):167-175.
    [43]Smith MR. Zoledronic acid to p revent skeletal comp lications in cancer: corroborating the evidence. Cancer Treat Rev,2005,31(supp 13):19-25.
    [44]Clohisy DR, Mantyh PW. Bone cancer pain and the role of RANKL/OPG. J Musculoskelet Neuronal Interact,2004,4(3):293-300.
    [45]Luger NM, Honore P, Sabino MAC, et al. Osteoprotegerin diminishes advanced bone cancer pain. Cancer Res,2001,61:4038-4047.
    [46]Honore P, Menning PM, Rogers SD, et al. Neurochemical plasticity in persistent inflammatory pain. Prog Brain Res,2000,129:357-363.
    [47]Honore P, Schwei J, Rogers SD, et al. Cellular and neurochemical remodeling of the spinal cord in bone cancer pain. Prog Brain Res,2000,129:389-397.
    [48]Fu SY, Gordon T. The cellular and molecular basis of peripheral nerve regeneration. Mol Neurobiol,1997,14:67-116.
    [49]Fukuoka T, Kondo E, Dai Y, et al. Brain-derived neurotrophic factor increases in the uninjured dorsal root ganglion neurons in selective spinal nerve ligation model. J Neurosci,2001,21:4891-4900.
    [50]Boucher TJ, McMahon SB. Neurotrophic factors and neuropathic pain. Curr Opin Pharmacol,2001,1:66-72.
    [51]Hoke A, Gordon T, Zochodne DW, et al. A decline in glial cell-line-derived neurotrophic factor expression is associated with impaired regeneration after long-term Schwann cell denervation. Exp Neurol,2002,173:77-85.
    [52]Zhu ZW, Friess H, diMola FF, et al. Nerve growth factor expression correlates with perineural invasion and pain in human pancreatic cancer. J Clin Oncol, 1999,17:2419-2428
    [53]Schneider MB, Standop J, Ulrich A, et al. Expression of nerve growth factors in pancreatic neural tissue and pancreatic cancer. J Histochem Cytochem,2001,49: 1205-1210.
    [54]Averill S, Michael GJ, Shortland PJ, et al. NGF and GDNF ameliorate the increase in ATF3 exp ression which occurs in dorsal root ganglion cells in
    response to peripheral nerve injury Eur Neurosci,2004,19 (6):1437-1445.
    [55]Halvorson KG, Kubota K, SevcikMA, et al. A blocking antibody to nerve growth factor attenuates skeletal pain induced by prostate tumor cells growing in bone. Cancer Res,2005,65 (20):9426-9435.
    [56]Halvorson KG, Kubota K, SevcikMA, et al. A blocking antibody to nerve growth factor attenuates skeletal pain induced by p rostate tumor cells growing in bone. Cancer Res,2005,65(20):9426-9435.
    [57]Mundy GR. Metastases to bone:causes, consequences, and therapeutic opportunities. Nat Rev Cancer,2002,2:584-593.
    [58]Mantyh PW, Clohisy DR, Koltzenburg M, et al. Molecular mechanisms of cancer pain. Nat Rev Cancer,2002,2:201-209.
    [59]O'Connell JX, Nanthakumar SS, Nielsen GP, et al. Osteoid osteoma:the uniquely innervated bone tumor. Mod Pathol,1998,11:175-180.
    [60]Seifert P, Spitznas M. Tumours may be innervated. Virchows Archiv,2001,438: 228-231.
    [61]Terada T, Matsunaga Y. S-100-positive nerve fibers in hepatocellular carcinoma and intrahepatic cholangiocarcinoma:an immunohistochemical study. Pathol Int, 2001,51:89-93
    [62]Mercadante S. Malignant bone pain:pathophysiology and treatment. Pain,1997, 69:1-18.
    [63]Millan MJ.The induction of pain:an integrative review.Prog Neurobiol. 1999,57:1-164.
    [64]Woolf CJ,Shortland P,Coggeshall RE.Peripheral nerve injury triggers central sprouting of myelinated afferents.Nature.1992,355:75-78.
    [65]Bleakman D, Alt A, Nisenbaum ES. Glutamate receptors and pain. Semin Cell Dev Biol,2006,17:592-604.
    [66]Quartaroli M, Carignani C, Dal Fomo G, et al. Potent antihyperalgesic activity without tolerance produced by glycine site antagonist of N-Methyl-D-Aspartate Receptor GV196771 A. Pharmacology,1999,290:158-169.
    [67]Willis WD. Role of neurotransmitters in sensitization of pain responses. Ann N Y Acad Sci,2001,933:142-156.
    [68]Lehre KP, Danbolt NC. The number of glutamate transporter subtype molecules at glutamatergic synapses:chemical and stereological quantification in young adult rat brain. Neurosci,1998,18:8751~8757.
    [69]Doubell TP, Mannion RJ, Woolf CJ. The dorsal horn:state dependent sensory processing, plasticity and the generation of pain. In:Wall PD, Melzack R, eds. Textbook of pain. London:Churchill Livingstone,1999:165-181.
    [70]Millan MJ.The induction of pain:an integrative review.Prog Neurobiol.1999, 57:1-164
    [71]Thomson LM,Zeng J, Terman GM. An N-methyl-D-aspartate receptor mediated large,low-f requency, spontant post synaptic current in neonatal rat spinal dorsal horn neurous.Neuroscience.2006.141 (3):1489-1501.
    [72]Bennett GJ. Update on the neurophysiology of pain transmission and modulation: focus on the NMDA-receptor. J Pain Symptom Manage,2000,19:S2-6.
    [73]Wang GM, Tian YK, Chen JP,et al. Evaluation of NR2B peptide as subunit vaccines against experimental neuropathic pain, Chinese Medical Journal,2007, 120(8):643-647.
    [74]ChizhBA, Headley PM, Tzschentke TM. NMDA receptor antagonists as analgesics focus on the NR2B subtype. Trend Pharmacol Sci,2001,22(12): 636-642.
    [75]Madry C, Mesic I, Bartholomaus I, et al. Principal role ofNR3 subunits in NR1/NR3 excitatory glycine recep tor function. Biochem Biophys ResCommun, 2007,354(1):102-108.
    [76]Dingledine R, Borges K, Bowie D, et al. The glutamate receptor ion channels. Pharmacol Rev,1999,51:7-61.
    [77]LoGrasso P, Mckelvy J. Advances in pain therapeutics CurrOp in Chem Biol, 2003,7(4) 452-456.
    [78]Jennifer M, Loftis, Aaron Janowsky. The N-methyl-D-aspartate receptor subunit NR2B localization, functional properties, regulation, and clinical implications. Pharmacology &Therapeutics,2003,97:55-85.
    [79]Blaise ML, Sowdhamini R, Rao MR, et al. Evolutionary trace analysis of ionotropic glutamate receptor sequences andmodeling the interactions of agonistswith differentNMDA receptor subunits. J Mol Model (On-line),2004, 10(5-6):305-316.
    [80]Chung HJ, Huang YH, Lau LF, et al. Regulation of the NMDA receptor complex and trafficking by activity-dependent phosphorylation of the NR2B subunit PDZ ligand. J Neurosci,2004,10; 24 (45):10248-10259.
    [81]Cais O, Sedlacek M, Horak M, et al. Temperature dependence of NR1-NR2B NMDA receptor channels. Neuroscience,2008,151(2):428-438.
    [82]Petrenko AB, Yamakura T, Baba H, et al. The role of N-methyl-D-aspartate (NMDA) receptors in pain:a review. Anesth Analg,2003,97(4):1108-1116.
    [83]Sabino MC, Ghilardi JR., Feia KJ, et al. The involvement of prostaglandins in tumorigenesis, tumor-induced osteolysis and bone cancer pain J Musculoskelet Neuronal Interact,2002,2(6):561-562.
    [84]Cain DM, Wacnik PW, Turner M, et al. Functional interactions between tumor and peripherall nerve:changes in excitability and morphology of primary afferent fibers in a murine model of cancer pain. J Neurosci,2001,21(23):9367-9376.
    [85]Yamamoto J, Kawamata T, Niiyama Y, et al. Down-regulationof mu opioid receptor expression within distinct subpopulations of dorsal root ganglion neurons in a murine model of bone cancer pain. Neuroscience,2008,151(3): 843-853.
    [86]Urch CE, Donovan-Rodriguez T, Dickenson AH. Alterations in dorsal horn neurones in a rat model of cancer-induced bone pain. Pain,2003,106(3): 347-356.
    [87]Meuser T, Pietruck C, Radbruch L, et al. Symptoms during cancer pain treatment following WHO-guidelines:A longitudinal follow-up study of symptom prevalence, severity and etiology. Pain,2001,93:247-257.
    [88]Masferrer JL, Leahy KM, Koki AT, et al. Antiangiogenic and antitumor activities of cyclooxygenase-inhibitors. Cancer Res,2000,60:1306-1311.
    [89]Kundu N, Yang Q, Dorsey R, et al. Increased cyclooxyge nase-2 (cox-2) expression and activity in a murine model of metastatic breast cancer. Int J Cancer,2001,93:681-686.
    [90]Moore BC, Simmons DL. COX-2 inhibition, apoptosis, and chemoprevention by nonsteroidal anti-inflammatory drugs. Curr Med Chem,2000,7:1131-1144.
    [91]Kurbel S, Kurbel B, Kovacic D, et al. Endothelin-secreting tumors and the idea of the pseudoectopic hormone secretion in tumors. Med Hypotheses,1999,52: 329-333.
    [92]Sabino MA, Ghilardi JR, Jongen JL, et al. Simultaneous reduction in cancer pain, bone destruction,and tumor growth by selective inhibition of cyclooxygenase-2. Cancer Res,2002,62(24):7343-7349.
    [93]Wacnik PW, Eikmeier LJ, Ruggles TR,et al. Functional interactions between
    tumor and peripheral nerve:morphology,algogen identification,and behavioral characterization of a new model of cancer pain J Neurosci,2001,21(23): 9355-9366.
    [94]Nelson JB, Carducci MA. The role of endothelin-1 and endothelin receptor antagonists in prostate cancer. BJU Int,2000,85(suppl 2):45-48.
    [95]Nelson JB, Hedican SP, George DJ, et al. Identification of endothelin-1 in the pathophysiology of metastatic adenocarcinoma of the prostate. Nat Med,1995, 1(9):944-999.
    [96]Pomonis JD, Rogers SD, Peters CM, et al. Expression and localization of endothelin receptors:implication for the involvement of peripheral glia in nociception. J Neurosci,2001,21:999-1006.
    [97]Dawas K, Laizidou M, Shankar A et al. Angiogenesis in cancer:the role of endothelin-1. Ann R Coll Surg Engl,1999,81:306-310.
    [98]Peters CM, Lindsay TH, Pomonis JD, et al. Endothelin and the tumorigenic component of bone cancer pain. Neuroscience,2004,126 (4):1043-1052.
    [99]Alanen K, Deng DX, Chakrabarti S, et al. Augmented exp ression of endothelin-1, endothelin-3 and the endothelin-B recep tor inbreast carcinoma. Histopathology,2000,36 (2):161-167.
    [100]Schmidt BL, Pickering V, Liu S, et al. Peripheral endothelin A receptor antagonism attenuates carcinoma induced pain. Eur Pain,2007,11 (4):406-414.
    [101]Sevcik MA, Luger NM, Mach DB, et al. Bone cancer pain:The effects of the bisphosphonate alendronate on pain, skeletal remodeling, tumor growth and tumor necrosis. Pain,2004,111:169-180.
    [102]Mach DB, Rogers SD, Sabino MC, et al. Origins of skeletal pain:Sensory and sympathetic innervation of the mouse femur. Neuroscience,2002,113:155-166.
    [103]Coleman RE. Bisphosphonates:Clinical experience. Oncologist,2004,9(Suppl 4):14-27.
    [104]Hiraga T, Williams PJ, Mundy GR, et al. The bisphosphonate ibandronate promotes apoptosis in MDA-MB-231 human breast cancer cells in bone metastases. Cancer Res,2001,61:4418-4424.
    [105]Vasikaran SD. Bisphosphonates:An overview with special reference to alendronate. Ann Clin Biochem,2001,38:608-623.
    [106]Neville-Webbe HL, Holen I, Coleman RE. The antitumour activity of bisphosphonates. Cancer Treat Rev,2002,28:305-319.
    [107]Olson TH, Riedl MS, Vulchanova L, et al. An acid sensing ion channel (ASIC) localizes to small primary afferent neurons in rats. Neuroreport,1998,9: 1109-1113.
    [108]Lesage P, Portenoy RK. Trends in cancer pain management. Cancer Control, 1999,6:136-145.
    [109]Tong D, Gillick L, Hendrickson FR. The palliation of symptomatic osseous metastases:Final results of the Study by the Radiation Therapy Oncology Group. Cancer,1982,50:893-899.
    [110]Hoskin PJ, Stratford MR, Folkes LK, et al. Effect of local radiotherapy for bone pain on urinary markers of osteoclast activity. Lancet,2000,355:1428-1429.
    [111]Goblirsch M, Mathews W, Lynch C, et al. Radiation treatment decreases bone cancer pain, osteolysis and tumor size. Radiat Res,2004,161:228-234.
    [112]Bennett MI, Simpson KH.Gabapentin in the treatment of neuropathic pain. Palliat Med,2004,18(1):5-11.
    [113]Donovan-Rodriguez T, Dickenson AH, Urch CE. Gabapentin normalizes spinal neuronal responses that correlate with behavior in a rat model of cancer-induced bone pain. Anesthesiology,2005,102(1):132-140.
    [114]Honore P, Mantyh PW. Bone cancer pain:From mechanism to model to therapy. Pain Med,2000,1:303-309.
    [115]Ghilardi JR, Rohrich H, Lindsay TH, et al. Selective blockade of the capsaicin receptor TRPV1 attenuates bone cancer pain. J Neurosci,2005,25:3126-3131.
    [116]Sevcik MA, Ghilardi JR, Peters CM, et al. Anti-NGF therapy profoundly reduces bone cancer pain and the accompanying increase in markers of peripheral and central sensitization. Pain,2005,115:128-141.
    [117]Sloane EM, Soderquist RG, Maier SF, Mahoney MJ, Watkins LR, Milligan ED. Long-term control of neuropathic pain in a non-viral gene therapy paradigm. Gene Ther.2009,16(4):470-475.
    [118]Meunier A, Pohl M. Lentiviral vectors for gene transfer into the spinal cord glial cells. Gene Ther.2009,16(4):476-482.
    [119]Wolfe D, Wechuck J, Krisky D, Mata M, Fink DJ. A Clinical Trial of Gene Therapy for Chronic Pain. Pain Medicine,2009,10(7):1325-1330.
    [120]Wolfe D, Mata M, Fink DJ. A human trial of HSV-mediated gene transfer for the treatment of chronic pain. Gene Ther,2009,16:455-460.
    [121]Marchand F, Jones NG, McMahon SB. Future treatment strategies for neuropathic pain. Handb Exp Pharmacol.2009(194):589-615.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700