针刺缓解慢性内脏痛敏的作用及其机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
慢性疼痛是现代医学发展和人类健康、保健所面临的严重问题之一,其中内脏疾患引起的慢性疼痛是临床上常见的疾病症候。内脏痛的发生部位深且定位模糊,同时伴有牵涉痛和植物神经反射等复杂的临床表现和特点。因此,人们对于内脏痛的认识远滞后于对躯体痛的了解,也使得慢性内脏痛的治疗缺乏特异性措施。
     众所周知,由于临床研究的局限性,动物实验研究成为研究内脏痛的重要手段。但是导致内脏痛研究滞后的一个重要方面就是缺乏较为理想的模拟人类慢性内脏痛的动物模型。2000年Al-Chaer等用大鼠成功地制作了一种新的质量较高的慢性内脏痛敏模型(IBS大鼠模型)。该模型的出现,为深入探讨慢性内脏痛的中枢机制提供了一个新的契机。
     研究表明,在内脏感觉敏感性增高状态下,机体对非伤害性刺激和伤害性刺激的处理,无论在外周还是在中枢水平都发生了改变。其中,中枢敏化的作用是研究热点之一,它是内脏痛发生发展过程中的关键性因素。有研究提示,如果抑制中枢敏化,则能有效地缓解慢性内脏痛。中枢敏化的形成涉及多种神经递质、受体系统,包括兴奋性氨基酸受体(EAA-R)、速激肽受体(NK-R)、5-羟色胺(5-HT)受体、α_2肾上腺素能受体、阿片受体等。其中兴奋性氨基酸递质受体系统在其中的作用引人注目。研究提示NMDA受体和non-NMDA受体均参与急性内脏伤害性感受,但究竟在慢性内脏痛疾病长期的病理演变过程中发挥了什么作用,这些与其在躯体痛中的作用有何不同和联系等,均值得进一步研究。
     国内外很多临床实践证实针刺对慢性内脏痛有着良好的治疗效果。但以往研究所利用的动物模型尚不理想。新的慢性内脏痛模型是否适用于针刺研究?如何利用该模型深化慢性内脏痛以及针刺治疗慢性内脏痛的机制探讨?对这些问题的解答,将为临床针刺治疗慢性内脏痛提供科学依据和理论基础,推动内脏痛的基础研究,促进针刺疗法不断走向世界。
     因此本项研究采用新的慢性内脏痛敏模型,并应用疼痛行为学观察、电生理细胞外记录和组织学技术对其进行评价;在该动物模型上,观察针刺对慢性内脏痛敏的治疗作用和作用规律,包括单次电针的即刻效应和多次电针的累加效应和不同针刺次数的治疗效应比较等;进而采用电生理细胞外记录等方法探讨脊髓兴奋性氨基酸递质受体系统在慢性内脏痛敏和针刺治疗慢性内脏痛敏中的作用。
Chronic pain originated from viscera is one of the most common symptoms in clinic and also is one of serious problems to be faced with in modern medicine development and human health care. Visceral pain occurs deeply and has complex clinical characteristics of diffuse and poorly localized, referred pain, autonomous nerve reflex and etc. Thus the understanding of viscera pain is far lagged behind that of somatic pain and less specific therapies of chronic visceral pain are valid.It is well known animal experiment is a very important method in the research of visceral pain owing to the limit of clinical study. While absence of ideal animal models mimicing human chronic visceral pain is a key aspect for the draggling of visceral pain. A new rat model of chronic visceral hyperalgesia was invented by Al-Chaer and his colleague in 2000, which provided a new approach for further exploration of the central mechanism of chronic visceral pain.It is reported that altered response to non-noxious and noxious stimulus happened in both peripheral and central levels under the state of increased visceral sensitivity. Central sensitization is one of hot spots in the study of visceral pain and it is a pivotal factor in the occurrence and progress of visceral pain. It is proved that chronic visceral pain could be effectively relieved if central sensitization was inhibited. Many systems of neurotransmitter and their receptors are involved in the forming and maintenance of central sensitization including systems of excitory amino acid receptor (EAA-R), tachykinin receptor (NK-R), 5-hydroxytryptamine (5-HT) receptor, α_2 adrenergic receptor, opiod receptor, etc. The role of EAA-R system in central sensitization of somatic pain is conspicuous and there is evidence shows that NMDA receptor and non-NMDA receptor take part in nociception of acute visceral pain, but what is the function of EAA-R system in the pathological development of chronic visceral pain and the relation and difference among them need further investigation.It has been proved by domestic and overseas practice in clinic that chronic visceral pain could be relieved by acupuncture treatment. But no ideal animal models were used in experimental study. Whether the new animal model could serve acupuncture research or not? How to make further exploration about the mechanism of chronic visceral pain and acupuncture analgesia? To answer the questions is to provide scientific evidence and academic theory of acupuncture
    against chronic visceral pain, to promote base research in visceral pain and also to advance acupuncture therapy to be more widely used in the world.New chronic visceral pain animal model will exert and behavioral pain reflex observation, electrophysiological extracellular recording and histological techniques will be used in the study. Then the therapeutic effect and the rule of the effect including instant effect of single treatment, cumulative effect of repetitive treatments and comparison of different effects of different treatments times will be observed. Finally the role of spinal EAA-R system in chronic visceral pain and therapeutic effect by acupuncture will be investigated by methods including electrophysiological extracellular recording.Results:1 Fabrication and evaluation of IBS rat model of chronic visceral hyperalgesia1.1 Fabrication of IBS rat modelMale neonatal SD rats (8th-21st postnatal) were given colorectal distention stimulation daily by using self-made balloons with reference to the method of Al-Chaer's. Evaluations were made when they became adults.1.2 Increased abdominal withdrawal reflex (AWR) score in IBS adult ratsSemi-quantitative score was given by observing AWR to graded colorectal distention (CRD) when the rats were awake. The results indicated that AWR of IBS group was significantly increased to graded CRD compared with that of normal rats. IBS rats manifested the sign of visceral hyperalgeisa.1.3 Increased discharge of abdominal electromyograms (AEMGs) in IBS ratsThe changes of AEMGs in response to graded CRD stimulation was recorded when the rats were under mild and stable anesthesia. The results indicated that AEMGs of IBS rats was abnormal increased in different intensities of CRD stimulation compared with that of normal rats, which reaffirm the occurrence of visceral hyperalgesia in IBS rats.1.4 Increased discharge of spinal wide dynamic range (WDR) neuron in IBS ratsThe changes of discharge of spinal WDR neuron in response to graded CRD stimulation was recorded when the rats were under anesthesia. The results indicated that discharge of WDR neuron was significantly increased in different intensities of CRD stimulation compared with that of normal rats, which suggested that the
    excitability of spinal neuron in IBS rats was obviously enhanced.1.5 Comparison of AWR score in IBS rats of 8th and 12th weeks oldThe changes of AWR score to graded CRD stimulation in IBS rats of 8th week old and 12th weeks old was observed to decide the persistence of visceral hyperalgesia. The results indicated that no significant difference between AWR in 8th IBS rats and that of 12th IBS rats, which suggested that visceral hyperalgesia could at least last when the rats were 12 weeks old.1.6 Comparison of diarrhea sign in IBS and normal adult ratsThe diarrhea sing was observed at the same day in adult IBS and normal rats. The result indicated that the incidence of diarrhea sign in IBS rats was distinctly higher to that of normal rats, which showed that the symptom of diarrhea in IBS rats was evident.1.7 The comparison of body weight in adult IBS and normal ratsThe body weight of adult IBS and normal rat (7th week old) was compared. The results indicated that no significant difference between body weight in adult IBS and normal rats, which implied that model fabrication itself had no influence on body weight.1.8 Stoll examination in adult IBS ratsThe stool of adult IBS and normal rats was examined and no abnormal changed in adult IBS rat was showed compared with that of adult normal rats.1.9 Histological examination of colorectal tissue in IBS ratsLocal histological changes were observed in adults IBS and normal rats. The results indicated that no abnormal infiltration of neutrophil and interstitial edema were observed by naked eye and microscope in adult IBS and normal rat.Summary:Obvious signs of visceral hyperalgesia were observed at peripheral and central levels by different methods in well-made adult IBS rats. The symptom of diarrhea was evident in IBS rat and the animal model belonged to functional chronic visceral hyperalgesia model. The behavior of IBS rats was stable and could last for long time. A new ideal animal model was provided for the research in the treatment of chronic visceral pain by acupuncture.2 Electro-acupuncture (EA) relived visceral hyperalgesia in IBS rats
    2.1 Single EA treatment decreased AWR score in IBS ratsThe instant effect (20-90 min after EA treatment stopped) of single EA on AWR score in IBS rats was observed. The rats were divided into three groups, model group (model), model plus EA treatment group (model + EA) and model plus sham EA treatment group (model + sham EA). The results indicated that AWR in response to graded CRD stimulation (20, 40, 60, 80 mmHg) of model + EA was significantly decreased compared with that in model group, while no change in group of model + sham EA. It suggested that single E A treatment could remarkably relieve visceral hyperalgesia in short time after EA stopped.2.2 Single EA treatment inhibited AEMGs discharge in IBS ratsThe instant effect (20-90 min after EA treatment stopped) of single EA on AEMGs discharge in IBS rats was observed. The rats were divided into three groups, model group (model), model plus EA treatment group (model + EA) and model plus sham EA treatment group (model + sham EA). The results indicated that discharge of AEMGs in response to graded CRD stimulation (20,40, 60, 80 mmHg) of model + EA was significantly inhibited compared with that in model group, while no change in group of model + sham EA. It again suggested that single EA treatment could remarkably suppress visceral hyperalgesia in short time after EA stopped.2.3 The effect of repeated EA treatments on AWR in IBS ratsThe effect of repeated EA treatments on AWR in IBS rats was observed. Repeated EA treatments were applied for 30 min every other day and lasted for 13 days. Consecutive assessments of AWR scores were conducted on the day when EA was not applied for 22 days. The results indicated that the effect of EA began 2 days after EA treatment (after one EA application), gradually increased to its maximum by the 8th day (4 EA applications) and lasted 5 days after the termination of EA treatment under different intensities of evoked CRD stimulations. Simultaneously, the effect of repeated sham EA treatments (needles were inserted into the acupoints similar to those in EA treatment but kept stilly without electrical stimulation) on AWR in IBS rats was observed. The results showed that no effect on AWR in IBS rats by sham EA treatments under different intensities of CRD stimulations. 2.4 The effect of repeated EA treatments on AEMGs in IBS ratsThe effect of repeated EA treatments on AEMGs in IBS rats was observed. Repeated EA treatments were applied for 30 min every other day and lasted for 13 days. Consecutive recording of AEMGs was conducted on the day when EA was not
    applied for 22 days. The results indicated that the effect of EA began 4 days after EA treatment (after two EA application), gradually increased to its maximum by the 8th day (4 EA applications) and lasted 5 days after the termination of EA treatment under different intensities of evoked CRD stimulations. Simultaneously, the effect of repeated sham EA treatments on AEMGs in IBS rats was observed. The results showed that no effect on discharge of AEMGs in IBS rats by sham EA treatments under different intensities of CRD stimulations.2.5 The effect of different times of EA treatments on AWR in IBS ratsThe effect of different times of EA treatments (1, 3, 5 and 7 EA treatments respectively) on AWR in IBS rats under the same CRD stimulus (60 mmHg) was observed. Repeated EA treatments were applied every other day and a consecutive assessment of AWR was conducted on the day when EA was not applied. Two to four extented AWR assessments were conducted after EA shopped. The results showed one time EA treatment could decrease AWR score to one day after EA shopped, three to five EA treatments could decrease AWR score to 3 days after EA shopped and 7 times of EA treatments could decrease AWR score to 5 days after EA shopped. It suggested that more EA treatments could prolong the post-effect after EA shopped.2.6 The effect of different times of EA treatments on AEMG in IBS ratsThe effect of different times of EA treatments (1, 3, 5 and 7 EA treatments respectively) on AEMGs in IBS rats under the same CRD stimulus (60 mmHg) was observed. Repeated EA treatments were applied every other day and consecutive recordings of AEMGs were conducted on the day when EA was not applied. Two to four prolonged AEMG recordings were conducted after EA shopped. The results showed one time EA treatment had no effect on discharge of AEMG, three to five EA treatments could inhibit AEMGs discharge to 3 days after EA shopped and 7 times of EA treatments could inhibit AEMGs discharge to 5 days after EA shopped. It again suggested that more EA treatments could prolong the post-effect after EA shopped.SummarySingle EA treatment could significantly relieve chronic visceral hyperalgesia. Cumulative effect of EA treatments was observed in the study, which began 2-4 days after EA treatment (after one or two EA applications), gradually increased to
    its maximum by the 8th day (4 EA applications) and lasted some days after the termination of EA treatment. The time of persistence of post-effect of repeated EA treatments was prolonged as more EA treatments applied. The results showed instant and cumulative effects of acupuncture on chronic visceral hyperalgesia and the therapeutic effects were lengthened as times of treatments increased.3 The role of spinal EAA-R system in visceral hyperalgesia and the therapeuticeffect of EA in IBS rats3.1 The effect of intrathecal injection of MK-801 on evoked discharge of spinal WDR neuron in IBS ratsThe effect of intrathecal injection of different dose of specific antagonist of NMDA receptor (MK-801) on evoked discharge of spinal WDR neuron in IBS rats was observed. Test for confirmation of cannula location was conducted 3 days after intrathecal cannulatation and experiments were done the next day. The base of evoked discharge of WDR neuron was recording first before injection. The rats were divide into four groups, low, medium and high dose of MK-801 (0.01 (ig, 0.1 jj.g and 1 ug respectively) and vehicle group. The results indicated that no effect on WDR neuron discharge in vehicle and low dose of MK-801 groups, injection of 0.1 ng MK-801 could significantly decrease WDR discharge 5 min after injection and the effect could last for 15 min, injection of 1.0 ug MK-801 could remarkably decrease WDR discharge and the effect could last for 45 min. The results suggested the i.t MK-801 could dose-dependently inhibit the evoked discharge of spinal WDR neuron to noxious stimulus.3.2 The effect of intrathecal injection of CNQX on evoked discharge of spinal WDR neuron in IBS ratsThe effect of intrathecal injection of specific antagonist of non-NMDA receptor (CNQX) on evoked discharge of spinal WDR neuron in IBS rats was observed. Test for confirmation of cannula location was conducted 3 days after intrathecal cannulatation and experiments were done the next day. The base of evoked discharge of WDR neuron was recording first before injection and then high dose of CNQX (100 ng) and vehicle were injected to investigate the effect on evoked WDR neuron discharge. The results indicated that no effect on WDR neuron discharge in vehicle and high dose of CNQX groups, which suggested i.t CNQX (100 ng) have no effect on the evoked discharge of spinal WDR neuron to noxious
    stimulus.3.3 The effect of EA on evoked discharge of spinal WDR neuron in IBS ratsThe effect of EA on evoked discharge of spinal WDR neuron in IBS rats was observed. Base discharge of evoked WDR neuron to noxious stimulus was firstly recorded and then the changes of evoked WDR neuron discharge were recorded during (30 min) and after EA treatment (240 min). The results indicated that the discharge of evoked WDR neuron was decreased 5 min after EA began, reached its maximum inhibition 20-30 min after EA treatment, gradually increased after EA stopped and no difference to base response to noxious stimulus 60 min after EA stopped. The results suggested that EA could significantly inhibited the evoked WDR neuron discharge to noxious stimulation and the maximum effect of EA was 20-30 min after EA treatment started.3.4 The effect of combination of EA treatment and intrathecal injection of MK-801 on evoked discharge of spinal WDR neuron in IBS ratsThe effect of combination of intrathecal injection of low dose of MK-801 and EA treatment on evoked discharge of spinal WDR neuron in IBS rats was observed. The rats were divided into four groups, EA group, MK-801 group (0.01 jxg), EA + MK-801 group and vehicle group. MK-801 was i.t. injected when EA began. The results showed no effect on WDR neuron discharge in vehicle and MK-801 groups while obviously suppressive effects in EA and EA + MK-801 groups, but no significant difference between EA and EA+ MK-801 groups. It suggested that low dose of MK-801 could not reinforce the inhibition of discharge of WDR neuron in IBS rats by acupuncture.SummaryThe results in the part indicated that spinal EAA-R system was involved in the chronic visceral hyperalgesia and NMDA receptor might mainly contributed to the maintenance of visceral hyperalgesia. EA could significantly inhibit the excitability of spinal WDR neurons, which maybe one possible mechanism of acupuncture against chronic visceral hyperalgesia.ConclusionsI. The well-made IBS model prettily mimiced the symptoms of clinical disease of IBS mainly including evident and stable visceral hyperalgesia, so it was an ideal
引文
[1] Cervero F, Laird J. M. A. Visceral pain. Lancet, 1999, 353: 2145-2148.
    [2] Harding J. E. Chronic pain: a disease in itself. Aust Nurses J, 1984, 14(4): 50-52.
    [3] Gallagher R. M. Chronic pain: a public health problem? Clin J Pain, 1998, 14(4): 277-279.
    [4] No authors listed. New frontier in medicine 'chronic pain, the disease'. RSDSA NEWS, 2002, Winter-Spring, 9(1) http://www.rsdsa-ca.org/Newsletter.htm.
    [5] Twillman R. K. Report from the 10th World Congress on Pain. J Pain Palliat Care Pharmacother, 2003, 17(2): 83-88.
    [6] Erdine S. Pain relief as a human right, Global Day Against Pain. IASP and EFIC initiative cosponsored by WHO, 11th October, 2004, Geneva.
    [7] 吕愈敏.肠道功能性疾病.郑芝田:《胃肠病学》,北京:人民卫生出版社.2000:730-735.
    [8] Pasricha EJ, Crowell M. D, Tougas G. Approach to the patient with abdominal pain. In: Yamada T, ed. Textbook of gastroenterology, 3rd ed Philadelphia: Lippincott Williams & Willkins, 1999: 795-815.
    [9] Olden K. W, Schuster M. M: Irritable bowel syndrome. In: Feldman M et al eds, Sleisenger and Fordtran's Gastrointestinal and liver disease, 6th ed, vol. 2, Philadelphia: WB Saunders, 1998: 1536-1547.
    [1O] 潘国宗,鲁素彩,柯美云,等.北京地区肠易激综合征的流行病学研究:一个整群、分层、随机的调查.中国流行病学杂志,2000,21:26-29.
    [11] No authors listed. Acupuncture. NIH Consensus Statement, 1997, Nov 3-5, 15(5): 1-34.
    [12] Yuan Y. Z, Tao R. J, Xu B, et al. Functional brain imaging in irritable bowel syndrome with rectal balloon-distention by using fMRI. World J Gastroenterol, 2003, 9(6): 1356-1360.
    [13] Sarkar S, Hobson A. R, Furlong P. L, et al. Central neural mechanisms mediating human visceral hypersensitivity. American Journal of Physiology-Gastrointestinal & Liver Physiology, 2001, 281(5): 1196-1202.
    [14] Petrenko A. B, Yamakura T, Baba H, et al. The role of N-methyl-D-aspartate (NMDA) receptors in pain. Anesthesia & Analgesia, 2003, 97(4): 1108-1116.
    [15] Ji Y, Traub R. J. Spinal NMDA receptors contribute to neuronal processing of acute noxious and nonnoxious colorectal stimulation in the rat. J Neurophysiol, 2001, (86): 1783-1791.
    [16] Castroman P. J, Ness T. J. Ketamine, an N-methyl-D-aspartate receptor antagonist, inhibits the reflex responses to distension of the rat urinary bladder. Anesthesiology, 2002, 96(6): 1401-1409.
    [17] Imamachi N, Saito Y, Hara, et al. The non-NMDA glutamate receptor antagonist CNQX augments lidocaine antinociception through a spinal action in rats. Anesthesia & Analgesia, 1999, 89(2): 416-421.
    [18] Jones J.C.(程鸿 主译)人类疾病动物模型.上海:上海医科大学出版社,1989:1-8.
    [19] 朱愉,多秀瀛.实验动物的疾病模型.天津:天津科技翻译出版公司,1997:3-5.
    [20] 苗明三.实验动物和动物实验技术.北京:中国中医药出版社,1997:3-7.
    [21] 施新猷.医学动物实验方法.北京:人民卫生出版社,1980:1-3.
    [22] 黄应堂.医学科学实验模型及评价方法.兰州:甘肃文化出版社,1998:1-5.
    [23] 杨斌,张励才,曾因明.内脏模型的研究进展.国外医学《麻醉学与复苏分册》,2003,24(2):101-103.
    [24] 江澄川,赵志奇,将豪.疼痛的基础与临床.上海:复旦大学出版社,上海医科大学出版社,2001:129-130.
    [25] Berthoud H. R, Neuhuber W. L. Functional and chemical anatomy of the afferent vagal system. Auton Neurosci, 2000, 85(1-3): 1-17.
    [26] Palecek J, Willis W. D. The dorsal column pathway facilitates visceromotor responses to colorectal distention after colon inflammation in rats. Pain, 2003, 104(3): 501-507.
    [27] 吕岩,李继硕,秦秉志,等.骶髓后连合核接受盆内脏伤害性信息传入的形态学证明.神经解剖学杂志,1996,12:201-208.
    [28] Ness T. J, Gebhart G. F. Visceral pain: a review of experimental studies, Pain. 1990, 41(2): 167-234.
    [29] 王宏琰,黄登凯.内脏痛的实验性研究.解剖学杂志,1995,18(3):282-287.
    [30] Kurihara T, Nonaka T, Tanabe T. Acetic acid conditioning stimulus Induces long-lasting antinociception of somatic inflammatory pain. Pharmacology, Biochemistry and Behavior, 2003, 74: 841-849.
    [31] Martinez V, Thakur S, Mogil J. S, et al. Differential effects of chemical and mechanical colonic Irritation on behavioral pain response to intrapedtoneal acetic acid in mice. Pain, 1999, 81: 179-186.
    [32] Miapamba M, Chery-Croze S, Detolle-Sarbach S, et al. Inflammation of the colonic wall Induced by formalin as a model of acute visceral pain. Pain, 1994, 57: 327-334.
    [33] Blair R. W, Ammons W. S, Foreman R. D. Response of thoracic spinothalamic and spinoreticular cells to coronary artery occlusion. J Neurophysiol, 1984, 51: 636-643.
    [34] Ness T. J, Gebhart G. F. A psychophysiological study in humans using phasic colonic distention as a noxious stimulus. Pain, 1990, 43: 377-386.
    [35] Ness T. J, Follett K. A. The development of tolerance to intrathecal morphine in rat models of visceral and cutaneous pain. Neuroscience letter, 1998, 22: 33-36.
    [36] Julia V, Mezzasalma T, Bueno L. Influence of bradykinin In gastrointestinal disorders and visceral pain Induced by acute or chronic Inflammation In rats. Dig Dis Sci, 1995, 40: 1913-1921.
    [37] Mayer E. A, Gebhart G. F. Basic and clinical aspects of visceral hyperalgesia. Gastroenterology, 1994; 107(1): 271-293.
    [38] AL-Chaer E. D, Traub R. J. Biological basis of visceral pain: recent developments. Pain, 2002, 96: 221-225.
    [39] Woolf C. J, Water E. T. Common patterns of plasticity contributing to nociceptive sensitization in mammals and aplysia. Trends Neurosci, 1991, 14: 74-78.
    [40] Fitzgerald M, Jennings E. The postnatal development of. spinal sensory processing. Proc Natl Acad Sci, 1999, 96: 7719-7722.
    [41] AL-Chaer E. D, Kawasaki M, Pasricha P. J. A new model of chronic visceral hypersensitivity in adult rats Induced by colon Irritation during postnatal development. Gastroenterology, 2000, 119: 1276-1285.
    [42] 吕愈敏.肠道功能性疾病.郑芝田,林三仁.《胃肠病学》(第三版).北京:人民卫生出版社,2000:730-731.
    [43] 张兵,张万岱.肠易激综合征.许国铭,李石.《现代消化病学》.北京:人民军医出版社,1999:860-862.
    [44] Gorard D. A, Farthing M. J. Intestinal motor function in irritable bowel syndrome. Dig Dis, 1994, 12(2): 72-76.
    [45] Snape W. J. Current concepts in the management of the irritable bowel syndrome. Rer Gastroentorol Mex, 1994, 59(2): 127-132.
    [46] Greenwood B, Rodriguez S, Decktor D, et al. Irritable bowel syndrome: a study to investigate the mechanisms of visceral hypersensitivity. J Okla State Med Assoc, 1996, 89(2): 47-50.
    [47] Naliboff B.D, Munakata J, Fullerton S, et al. Evidence for two distinct perceptual alterations in irritable bowel syndrome. Gut, 1997, 41(4): 505-512.
    [48] Mertz H.R. New concepts of irritable bowel syndrome. Curr Gastroenterol Rep, 1999, 1(5): 433-440.
    [49] Bonaz B. Visceral sensitivity perturbation integration in the brain-gut axis in functional digestive disorders. J Physiol Pharmacol, 2003, 54(Suppl 4): 27-42.
    [50] Koloski N.A, Talley N.J, Boyce P.M. Irritable bowel syndrome in women: the physician-patient relationship evolving. J Am Osteopath Assoc, 2001,101(12 Suppl Pt 2):S12-S16.
    [51]高峻,李兆申.肠易激综合征的病因研究进展.国外医学.消化系统分册, 2002, 22(2): 87-89.
    [52] Spiller R.C, Jenkins D, Thornley J.P, et al. Increased rectal mucosal enteroendocrine cells, T lymphocytes, and increased gut permeability following acute Campylobacter enteritis and in post-dysenteric irritable bowel syndrome. Gut, 2000, 47(6): 804-811.
    [53] Pimentel M, Chang M, Chow E.J, et al. Identification of a prodromal period in Crohn's disease but not ulcerative colitis. Am J Gastroenterol, 2000, 95(12): 3458-3462.
    [54] Ali A, Toner B.B, Stuckless N, et al. Emotional abuse, self-blame, and self-silencing in women with irritable bowel syndrome. Psychosom Med, 2000, 62(1): 76-82.
    [55] Fass R, Fullerton S, Tung S, et al. Sleep disturbances in clinic patients with functional bowel disorders. Am J Gastroenterol, 2000, 95(5): 1195-2000.
    [56] Naliboff B.D, Derbyshire S.W, Munakata J, et al. Cerebral activation in patients with irritable bowel syndrome and control subjects during rectosigmoid stimulation. Psychosom Med, 2001, 63(3): 365-375.
    [57] Talley N.J, Howell S, Poulton R. The irritable bowel syndrome and psychiatric disorders in the community: is there a link? Am J Gastroenterol, 2001, 96(4): 1072-1079.
    [58] Beverley GV.M, Matthew G, Wesley G, et al. Stereotaxic delivery of corticosterone to the amygdala modulates colonic sensitivity in rats. Brain Res, 2001,893(1-2): 135-142.
    [59] Locke G. R, Zinsmeister A. R, Talley N. J, et al. Familial association in adults with functional gastrointestinal disorders. Mayo Clin Proc, 2000, 75: 907-912.
    [60] Andrews K, Fitzgerald M. The cutaneous withdrawal reflex in human neonates: sensitization, receptive fields, and effects of contralteral stimulation. Pain, 1994, 56: 95-101.
    [61] Fitzgerald M. Development of pain mechanisms. Br Med Bull, 1991, 47: 667-675.
    [62] Sandkuhler J, Liu X. G.. Induction of long-term potentiation at spinal synapses by noxious stimulation or nerve injury. European Journal of Neuroscience, 1998, 10(7): 1446-1460.
    [63] Ikeda H, Heinke B, Ruscheweyh R, et al. Synaptic plasticity in spinal lamina Ⅰ projection neurons that mediate hyperalgesia. Science, 2003, 299: 1237-1240.
    [64] No authors listed, http://www.cutech.edu.cn/zhoukan2003/zk03_57_209.asphtml 2003-02-21/2005-04-14.
    [65] Curatolo M, Arendt-Nielsen L, Petersen-Felix S. Evidence, mechanisms, and clinical implications of central hypersensitivity in chronic pain after whiplash injury. Clin J Pain, 2004, 20(6): 469-476.
    [66] Rosenow J. M, Henderson J. M. Anatomy and physiology of chronic pain. Neurosurg Clin N Am, 2003, 14(3): 445-462.
    [67] 钟敏,张励才,曾因明.痛觉中枢敏化的分子机制.国外医学.麻醉学与复苏分册,2000,21(3):148-151.
    [68] Mayer E. A, Gebhart G. F. Basic and clinical aspects of visceral hyperalgesia. Gastroenterology, 1994, 107: 271-293.
    [69] Robinson D, Calejesan A. A, Zhuo M. Long-lasting changes in rostral ventral medulla neuronal activity after inflammation. J Pain, 2002, 3(4): 292-300.
    [70] Benani A, Vol C, Heurtaux T, et al. Up-regulation of fatty acid metabolizing-enzymes mRNA in rat spinal cord during persistent peripheral local inflammation. Eur J Neurosci, 2003, 18(7): 1904-1914.
    [71] Ruscheweyh R, Sandkuhler J. Opioids and central sensitisation: Ⅱ. Induction and reversal of hyperalgesia. Eur J Pain, 2005, 9(2): 149-152.
    [72] Sun R. Q, Tu Y. J, Lawand N. B, et al. Calcitonin gene-related peptide receptor activation produces PKA- and PKC-dependent mechanical hyperalgesia and central sensitization. J Neurophysiol, 2004, 92(5): 2859-2866.
    [73] Sivilotti L, Woolf C. J. The contribution of GABAA and glycine receptors to central sensitization: disinhibition and touch-evoked allodynia in the spinal cord. J Neurophysiol, 1994, 72(1): 169-179.
    [74] Willis W. D. Role of neurotransmitters in sensitization of pain responses. Ann N Y Acad Sci, 2001, 933: 142-156.
    [75] 许绍芬.兴奋性氨基酸类递质.许绍芬.《神经生物学》(第二版),上海:上海医科大学出版社,1999:202-208.
    [76] Moloney M. G. Excitatory amino acids. Nat Prod Rep, 2002, 19(5): 597-616.
    [77] Soliman A. C, Yu J. S, Coderre T. J. mGlu and NMDA receptor contributions to capsaicin-induced thermal and mechanical hypersensitivity. Neuropharrnacology, 2005, 48(3): 325-332.
    [78] Zhang L, Lu Y, Chen Y, et al. Group Ⅰ metabotropic glutamate receptor antagonists block secondary thermal hyperalgesia in rats with knee joint inflammation. J Pharmacol Exp Ther, 2002, 300(1): 149-156.
    [79] 阮祥才,佘守章.兴奋性氨基酸与疼痛.国外医学·麻醉学与复苏分册,2000,21(3):151-153.
    [80] Bennett G. J. Update on the neurophysiology of pain transmission and modulation: focus on the NMDA-receptor. J Pain Symptom Manage, 2000, 19(1 Suppl): S2-S6.
    [81] Schaible H. G, Ebersberger A, Von Banchet G. S. Mechanisms of pain in arthritis. Ann N Y Acad Sci, 2002, 966: 343-354.
    [82] Sang C. N. NMDA-receptor antagonists in neuropathic pain: experimental methods to clinical trials. J Pain Symptom Manage, 2000, 19(1 Suppl): S21-S25.
    [83] Coderre T. J, Melzack R. The contribution of excitatory amino acids to central sensitization and persistent nociception after formalin-induced tissue injury. J Neurosci, 1992, 12: 3665-3670.
    [84] Dickenson A. H, Sullivan A. F. Differential effects of excitatory amino acid antagonists on dorsal horn nociceptive neurones in the rat. Brain Res, 1990, 506: 31-39.
    [85] Dickenson A. H, Sullivan A. F. Evidence for a role of the NMDA receptor in the frequency dependent potentiation of deep rat dorsal horn nociceptive neurones following C fibre stimulation. N europharmacology, 1987, 26: 1235-1238.
    [86] Haley J. E, Sullivan A. F, Dickenson A. H. Evidence for spinal N-methyl-D-aspartate receptor involvement in prolonged chemical nociception in the rat. Brain Res, 1990, 518: 218-226.
    [87] Nishiyama T. Interaction among NMDA receptor-, NMDA glycine site- and AMPA receptor antagonists in spinally mediated analgesia. Can J Anaesth, 2000, 47: 693-698.
    [88] Olivar T, Laird J.M. Differential effects of N-methyl-D-aspartate receptor blockade on nociceptive somatic and visceral reflexes. Pain, 1999, 79: 67-73.
    [89] McRoberts J.A, Coutinho S.V, Marvizon J.C, et al. Role of peripheral N-methyl-D-aspartate (NMDA) receptors in visceral nociception in rats. Gastroenterology, 2001,120(7): 1737-1748.
    [90] Zhai Q.Z, Traub R.J. The NMDA receptor antagonist MK-801 attenuate c-Fos expression in the lumbosacral spinal cord following repetitive noxious and non-noxious colorectal distention. Pain, 1999, 83: 321-329.
    [91] Ji Y, Traub R.J. Spinal NMDA receptors contribute to neuronal processing of acute noxious and non-noxious colorectal stimulation in the rat. J Neurophysiol, 2001,86: 1783-1791.
    [92] Willeft R.P, Woolf C.J, Hobson A.R, et al. The development and maintenance of human visceral pain hyperalgesia is dependent on the N-Methyl-D-Aspartate receptor, gastroenterology, 2004,126: 683-692.
    [93] Al-Chaer E.D, Lawand N.B, Westlund K.N, et al. Pelvic visceral input into the nucleus gracilis is largely mediated by the postsynpatic dorsal column pathway. J Neurophysiol, 1996, 76: 2675-2690.
    [94] Coutinho S.V, Meller S.T, Gebhart G.F. Intracolonic zymosan produces visceral hyperalgesia in the rat that is mediated by spinal NMDA and non-NMDA receptors. Brain Res, 1996,736:7-15.
    [95] Ide Y, Maehara Y, Tsukahara S, et al. The effects of an intrathecal NMDA antagonist (AP5) on the behavioral changes induced by colorectal inflammation with turpentine in rats. Life Sci, 1997, 60: 1359-1363.
    [96] Song X.J. Zhao Z.Q. Involvement of NMDA and non-NMDA receptors in transmission of spinal visceral nociception in cat. Acta Pharmacologica Sinica, 1999, 20(4): 308-312.
    [97] Eide P.K. Wind-up and the NMDA receptor complex from a clinical perspective. Eur J Pain, 2000, 4(1): 5-15.
    [98] Aanonsen L.M, Lei S.Z. Excitatory amino acid receptors and nociceptive neurotransmission in rat spinal cord. Pain, 1990, 41: 309-321.
    [99] Tao Y.X, Zhao Z.Q. NMDA receptors mediate formalin-induced expression of
     c-fos in rat spinal dorsal horn. Acta Pharmacol Sinica, 1998, 19: 506-508.
    [100] Kangraga I, Randic M. Outflow of endogenous aspartate and glutamate from the spinal dorsa horn in vitro by activation of low- and high- threshold primary afferent fibers, modulation by μ opiods. Brain Res, 1991, 553: 347-352.
    [101] 赵志奇. 疼痛及其脊髓机理. 上海:上海科技教育出版社, 2000: 126-131.
    [102] Coutinho S.V, Urban M.O, Gebhart G.F. Role of glutamate receptors and nitric oxide in the rostral ventromedial medulla in visceral hyperalgesia. Pain, 1998, 78(1): 59-69.
    [103] Imamachi N, Saito Y, Hara K, et al. The non-NMDA glutamate receptor antagonist CNQX augments lidocaine antinociception through a spinal action in rats. Anesth Analg, 1999, 89(2): 416-421.
    [104] Ji Y, Traub R.J. Differential effects of spinal CNQX on two populations of dorsal horn neurons responding to colorectal distension in the rat. Pain, 2002, 99(1-2): 217-222.
    [105] Traub R.J, Pechman P, Iadarola M.J, et al. Fos-like proteins in the lumbosacral spinal cord following noxious and non-noxious colorectal distention in the rat. Pain, 1992, 49(3): 393-403.
    [106] Traub R.J, Murphy A. Colonic inflammation induces fos expression in the thoracolumbar spinal cord increasing activity in the spinoparabracbial pathway. Pain, 2002, 95(1-2): 93-102.
    [107] Ness T.J, Gebhart GF. Acute inflammation differentially alters the activity of two classes of rat spinal visceral nociceptive neurons, Neuroscience Letters. 2000, 281:131-134.
    [108] Neugebauer V, Lucke T, Grubb B, et al. The involvement of N-methyl-D-aspartate (NMDA) and non-NMDA receptors in the responsiveness of rat spinal neurons with input from the chronically inflamed ankle. Neurosci Lett, 1994, 170(2): 237-240.
    [109] Neugebauer V, Lucke T, Schaible H.G. N-methyl-D-aspartate (NMDA) and non-NMDA receptor antagonists block the hyperexcitability of dorsal horn neurons during development of acute arthritis in rat's knee joint. J Neurophysiol, 1993, 70(4): 1365-1377.
    [110] Wood P.L. The NMDA receptor complex: a long and winding road to therapeutics. Idrugs, 2005, 8(3): 229-235.
    [111] McRoberts J.A, Coutinho S.V, Marvizon J.C, et al. Role of peripheral
     N-methyl-D-aspartate (NMDA) receptors in visceral nociception in rats. Gastroenterology, 2001, 120(7): 1737-1748.
    [112] Wang C. Wang Y. Zhao Z. Peripheral NMDA and non-NMDA receptors contribute to nociception: an electrophysiological study. Brain Research Bulletin, 2000, 52(1): 31-34.
    [113] Salter M. W. Cellular neuroplasticity mechanisms mediating pain persistence. J Orofac Pain, 2004, 18(4): 318-324.
    [114] Sato E, Takano Y, Kuno Y, et al. Involvement of spinal tyrosine kinase in inflammatory and N-methyl-D-aspartate-induced hyperalgesia in rats. Eur J Pharmacol, 2003, 468(3): 191-198.
    [115] Coutinho S. V, Urban M. O, Gebhart G. F. The role of CNS NMDA receptors and nitric oxide in visceral hyperalgesia. Eur J Pharmacol, 2001, 429(1-3): 319-325.
    [116] Riedel W, Neeck G. Nociception, pain, and antinociception: current concepts. Z Rheumatol, 2001, 60(6): 404-415.
    [117] 王德深.针灸基础知识.北京:科学出版社,1985:1-8.
    [118] 张笑平.针灸作用机理研究.合肥:安徽科学技术出版社,1983:1-12.
    [119] 王德深.中国针灸学.天津:天津科技翻译出版公司,1992:7-53.
    [120] 唐浩.针灸与脏腑经络的整体观.光明中医杂志,1995,3:19-20.
    [121] 刘乡.张守信.魏北桦.穴位相对特异性与经络或神经节段关系的实验研究.针刺研究,1995,20(1):54-59.
    [122] 上海中医学院编.针灸学.北京:人民卫生出版社,1974:1-20.
    [123] 翁恩琪.痛与镇痛.上海:科学技术出版社,1987:182-188.
    [124] Zhang Y. Q, Ji G. C, Wu G. C, et al. Kynurenic acid enhances electroacupuncture analgesia in normal and carrageenan-injected rats. Brain Res, 2003, 966(2): 300-307.
    [125] Zhang Y. Q, Ji G. C, Wu G. C, et al. Excitatory amino acid receptor antagonists and electroacupuncture synergetically inhibit carrageenan-induced behavioral hyperalgesia and spinal fos expression in rats. Pain, 2002, 99(3): 525-535.
    [126] Wang S. J, Omori N, Li F, et al. Enhanced expression of phospho-Akt by electro-acupuncture in normal rat brain. Neurological Research, 2002, 24(7): 719-724.
    [127] de Medeiros M. A, Canteras N. S, Suchecki D, et al. Analgesia and c-Fos expression in the periaqueductal gray induced by electroacupuncture at the Zusanli point in rats. Brain Research, 2003, 973(2): 196-204.
    [128] Ting H, Liao J. M, Lin C. F, et al. Pressor effect on blood pressure and renal nerve activity elicited by electroacupuncture in intact and acute hemorrhage rats. Neuroscience Letters, 2002, 327(1): 5-8.
    [129] Chiu J. H, Chung M. S. Cheng H. C, et al. Different central manifestations in response to electroacupuncture at analgesic and nonanalgesic acupoints in rats: a manganese-enhanced functional magnetic resonance imaging study. Canadian Journal of Veterinary Research, 2003, 67(2): 94-101.
    [130] Cui K. M, Li W. M, Gao X, et al. Electro-acupuncture relieves chronic visceral hyperalgesia in rats. Neuroscience Letters, 2005, 376 (1): 20-23.
    [131] 张兆发,庄鼎.电针基础与临床.北京:中国科学技术出版社,1993:7-51.
    [132] 朱兵.弥散性伤害抑制性控制机制.江澄川,赵志奇,将豪.主编《疼痛的基础与临床》.上海:复旦大学出版社,上海医科大学出版社,2001,137-149.
    [133] Bing Z, Villanueva L, Le B. D. Acupuncture and diffuse noxious inhibitory controls: naloxone reversible depression of activities of trigeminal convergent neurons. Neuroscience, 1990, 37: 809-816.
    [134] 韩济生.针刺镇痛原理.上海:上海科技教育出版社,1999:112-125.
    [135] Ulett G. A, Han S, Han J. S. Electroacupuncture: mechanisms and clinical application. Biol Psychiatry, 1998, 44(2): 129-138.
    [136] 王贺春,万有,姚磊,等.不同频度电针治疗大鼠慢性神经源性痛的疗效比较.针刺研究,2002,27(2):112-118.
    [137] 吴鎏桢,崔彩莲,韩济生.多次电针刺激抑制大鼠吗啡戒断症状的累加效应及长时程后效应。中国疼痛医学杂志,2001,7(2):105-108.
    [138] 罗非,李桶,田津斌,等.反复电针对单发佐剂关节炎模型大鼠脊髓P物质、八肽胆囊收缩素及甲硫氨酸脑啡肽样免疫活性物质自发释放的影响.中国疼痛医学杂志,1996,2(3):169-175.
    [139] 吴根诚,曹小定.痛与镇痛.许绍芬.《神经生物学》(第二版),上海:上海医科大学出版社,1999:345-349.
    [140] Kitade T. Odahara Y. Shinohara S. et al. Studies on the enhanced effect of acupuncture analgesia and acupuncture anesthesia by D-phenylalanine (2nd report)-schedule of administration and clinical effects in low back pain and tooth extraction. Acupuncture & Electro-Therapeutics Researc, 1990, 15(2): 121-135.
    [141] 张秀琳,朱崇斌,吴根诚,等.大鼠侧脑室及鞘内注射孤啡肽减弱电针镇痛效应.解放军医学高等专科学校学报,1996,24(4):12-15.
    [142] 谢虹,马飞,高秀,等.曲马多加强针刺对关节炎大鼠的镇痛作用.针刺

© 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