用户名: 密码: 验证码:
IL-12、TNF-α及其受体在AIDP、AMAN中的作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
本文以研究IL-12、TNF-α及其相应受体在AIDP和AMAN发病中的作用为目的,探讨其可能的发病机制,为抗炎症细胞因子药物治疗GBS提供理论依据。依据GBS患者电生理的改变将患者分为AIDP组和AMAN组,根据患者是否接受了IVIg治疗,将患者分为未经IVIg治疗AIDP组、未经IVIg治疗AMAN组、IVIg治疗AIDP组、IVIg治疗AMAN组,应用FACS检测各组患者体内淋巴细胞、单核细胞表面IL-12Rβ1、IL-12Rβ2、TNFR1、TNFR2及细胞内部IL-12和TNF-α表达量的经时变化,并应用ELISA方法同时检测各组患者血浆中IL-12P40、IL-12P70、TNF-α及其可溶性受体sTNFR1、sTNFR2含量。分析炎症细胞因子及其受体表达与临床症状及IVIg治疗之间的关系。FACS检测结果发现,未经IVIg治疗AIDP患者在急性期IL-12、IL-12Rβ1水平增高,恢复期TNF-α和TNFR1水平增高,未经IVIg治疗AMAN患者急性期TNF-α水平增高;IVIg治疗AIDP组患者TNFR1及IL-12Rβ1表达量下降,IVIg治疗AMAN组患者TNF-α和TNFR2表达量增加;ELISA检测结果发现,IVIg治疗后AIDP组患者血浆中可溶性受体sTNFR1和IL-12P70含量下降。研究结果提示IL-12及IL-12Rβ1参与了AIDP的发病,在疾病发展中起促进作用,而AMAN组没发现类似的变化。TNF-α通过与不同的受体结合对GBS发挥着双向调节作用,在AIDP急性期TNF-α主要通过与TNFR1结合来实现其破坏性作用,在AMAN恢复期,TNF-α主要通过与TNFR2结合来发挥其保护性作用。IVIg通过下调TNFR1和IL-12Rβ1促进了AIDP的恢复,通过上调TNF-α和TNFR2促进了AMAN的恢复。
Guillain-Barrésyndrome (GBS) is an immune-mediated disease of the peripheral nerves involving both the myelin sheath and axons. Among its four subtypes of acute peripheral neuropathy, the most common is acute inflammatory demyelinating polyradiculoneuropathy (AIDP); another common subtype is acute motor axonal neuropathy (AMAN). GBS is generally accepted as a post-infectious autoimmune disease and is involved in molecular mimicry trigged by many antecedent pathogens. Numerous inflammatory cytokines and chemokines are thought to play pivotal roles in initiating, enhancing and perpetuating pathphysiological procedure in GBS.
     Interleukin 12 (IL-12) is an important candidate cytokine for the study of autoimmune disease because of its proinflammatory role with potent immune-regulatory activities and pivotal place in determining the differentiation and generation of Th1 cell. In previous studies, we reported that up-regulated IL-12 mRNA expression was found at the height of experimental autoimmune neuritis (EAN) course and IL-12p40 deficient mice showed significantly less severity of EAN, a widely accepted animal model for GBS in humans. However, whether IL-12 is implicated in the pathogenesis of GBS and its main subtypes (AIDP and AMAN) is still not clear.
     As another key mediator of regulatory processes in autoimmune demyelinating diseases, tumour necrosis factor-α(TNF-α) is considered crucial in the pathogenesis of GBS.The function of TNF-αwas initially characterized as inflammatory activity; however it was later found to be pleiotropic, exemplified by its anti-inflammatory effect. The contradictory biological activities of TNF-αare due to binding to its two receptors (TNFR1 and TNFR2) with distinct effects.
     In addition, as an effective immune-modulating therapy, intravenous high dose immunoglobulin (IVIg) is replacing corticosteroids in GBS. Hughes and his colleagues reported IVIg improved the disability grade scale in five trials with altogether 582 participants. The efficiency of immunoglobulin may at least partially be explained by the participation of modulating cytokines and their receptors in immunoglobulin therapy; however, the exact mechanism of IVIg remains unclear.
     In this study, we hypothesized that TNF-αand IL-12 molecules, as well as their receptors might play the different roles in the subtypes of GBS since AIDP and AMAN differ with regard to clinical pattern, electrophysiological testing, pathology and pathogenesis. We also observed the effects of IVIg on expression of TNF-αand IL-12 molecules, as well as their receptors in AIDP and AMAN.
     Materials and Methods
     Patients and controls
     We recruited 28 patients fulfilling international diagnostic criteria for GBS or its variants from the Department of Neurology, the First Hospital, Jilin University, Changchun, China, and 13 healthy controls between December 2006 and August 2007. All patients were classified neurophysiologically as AMAN (n=18) and AIDP (n=10), using motor nerve conduction criteria. Severity of GBS was scored by the use of Hughes degree (a functional disability scale). The pre-treatment patients were defined as absent of any immune-modulating drugs within 3 months and other treatments within 3 months and the post-treatment patients were treated with IVIg at a dose of 0.4g/kg body weight per day for 5 days consecutively. Blood was sampled two times to observe the different course of GBS, at acute phase (1-14 days from onset day) and at plateau phase (15-32 days from onset day). Patients with chronic inflammatory demyelinating polyneuropathy were excluded. The clinical characteristics of all subjects are shown in Table 1. Thirteen healthy donors (7 females and 6 males, age 21-58 years) were included. The present study was approved by the Human Ethics Committee of Jilin Province, China and informed consent was obtained from all patients and healthy controls.
     Peripheral blood mononuclear cells preparation and flow cytometric analysis
     Blood samples were taken at 15:00 pm each day. Lymphoprep (Axis-Shield PoC AS, Oslo, Norway) gradient centrifugation was used to separate peripheral blood mononuclear cells (PBMCs). Mononuclear cells were washed twice in PBS and cell viability measured by Trypan blue exclusion was confirmed to exceed 95%. Serum samples were also collected and cryopreserved at -80°C for further ELISA performance. An amount of 1000 units of protease inhibitor (Aprotinin, Sigma, London, UK) were used to prevent protein degradation.
     Three-colour flow cytometric technique was used in the analysis of surface phenotypes of PBMCs and cytokine as well as their receptor expression. Briefly, 106 cells/ml were incubated with the following PE- or FITC-conjugated anti-human monoclonal antibodies (mAbs): anti-CD3 (marker for T cells) and anti-CD14 (marker for monocytes, Caltag Laboratories, Burlingame, California, USA); anti-TNF-αreceptor 1 (TNFR1, CD120a; Serotec, Oxford, UK), anti-TNFR2 (CD120b; Caltag), anti-IL-12RΒ1 and IL-12R2 (BD Biosciences Pharmingen, San Diego, CA); moreover APC-conjugated mAb anti-TNF-αand anti-IL-12 (p40/p70) (BD) were used with the corresponding isotype controls. Cells were incubated for 20 min at room temperature and subsequently fixed in 2% paraformaldehyded PBS and stored at 4℃until flow cytometric analysis by an FACSCalibur cytometer using CellQuest software (Becton Dickinson, San Jose, California, USA). The molecule expression was assessed by determining of the positive percentage of the PBMCs labelled with fluorescent mAbs directed against the respective molecules.
     ELISA measurements of soluble TNF-αand its receptors, as well as IL-12p40/70
     To measure the levels of serum TNF-α, soluble TNFR1 and TNFR2, IL-12p40 and IL-12p70 ELISA was performed according to the manufacturer’s instructions. The serum samples were all diluted 1:10 for TNF-α(Diaclon, Becancon, France), TNFRs (Bender MedSystem Inc., Vienna, Austria) and IL-12p40/70 (BD) detection. The assays were carried out in 96-well plates, measured spectrophotometrically at 450 nm (Molecular Devices Spectra MAX 250). All assays were done in duplicate. Statistical analysis
     Data were expressed as the mean±SD. For statistical analysis, differences of mean values were tested with one way analysis of variance (ANOVA) test for multiple comparisons and the student-t test for two groups, using SPSS software (version 11.5). Reported P-values are two-tailed and considered statistically significant at P < 0.05. Spearman rank test was used for correlation analysis.
     Results
     Clinical profiles
     The clinical features of 28 patients with GBS are shown in Table 1. In total 21 patients had the antecedent infection; namely, 7 with gastrointestinal infection, 9 with upper respiratory tract infection, 4 with gastrointestinal and upper respiratory tract infection and 1 with chickenpox infection. After intravenous immunoglobulin therapy, based on the Hughes degree during acute (named T1) and plateau phases (named T2), the clinical severity remitted during plateau phase in 7 out of 11 (63.6%) patients with AIDP and 4 out of 10 (40%) patients with AMAN. On the other hand, only 1 out of 4 untreated patients with AIDP and none with
     AMAN were found remitted during plateau phase.
     Increased IL-12, IL-12Rβ1 in AIDP and TNF-αin AMAN during the acute phase, and increased TNF-αand TNFR1 during the plateau phase of AIDP
     To explore the role of TNF-αand IL-12 as well as their receptors in the pathogenesis of AIDP and AMAN, we examined these molecules’expression on circulating T cells (CD3) and monocytes (CD14) of AIDP and AMAN without any treatment at the acute and plateau phases, respectively. Our data showed that in acute phase of AIDP, the expressions of IL-12Rβ1 clearly increased on T cells when compared with health controls (P< 0.05). A higher level of IL-12 was found in acute phase of AIDP than in plateau (P< 0.05). The levels of TNF-α(P< 0.01) and TNFR1 (P< 0.05) were also found markedly up-regulated during plateau phase in AIDP compared with health controls. Thus, increased levels of IL-12 and IL-12Rβ1 correlated directly with disease severity of AIDP and reduced their expression in parallel with clinical recovery in AIDP patients. However, the role of TNF-αin AIDP seems the reverse compared to IL-12 at the plateau phase of disease. Interestingly in AMAN, a higher level of TNF-αwas observed on both T cells and monocytes (compared with health controls) in acute phase (P< 0.05). This suggests that TNF-αappears to contribute AMAN development due to an inherent biological activity change of TNF-αin AMAN. IVIg therapy altered the profiles of TNF-αand its receptors, as well as IL-12RΒ1 expression, on cells
     To explore the mechanisms of the therapeutic efficiency of IVIg in GBS, we observed the profiles of TNF-α, IL-12 and their receptors’expression on circulating immune cells before and after IVIg treatment at acute and plateau phases of the patients by FACS. The results of FACS were estimated as the positive percentage, i.e. the above molecules’expression on T cells and monocytes in patients.
     In plateau phase of AIDP group, there was significant decrease in the percentage of TNFR1 (P< 0.05) and IL-12Rβ1 (P< 0.05) expression on monocytes between before and after treatment with IVIg. Interestingly, when compared with healthy controls, the level of IL-12Rβ1 on the circulating pre-treated monocytes was much higher (P< 0.05); whereas after treatment, IL-12Rβ1 expression decreased at a low level, dramatically less than healthy controls. Additionally, IVIg treatment did not alter markedly the expression of other molecules in AIDP either at acute or plateau phases.
     For confirming our findings in AIDP, the correlations between these molecules expression and clinical severity as well as courses were analyzed according to the full neurological assessment (Hughes RA, 1978). Our results indicated that the clinical severity in the nine patients with AIDP was associated significantly with high expression of IL-12Rβ1 in the acute phase (P< 0.05) and TNFR1 in the plateau phase (P< 0.05) on T cells. There were no significant correlations between expression of other molecules and clinical profile.
     In acute phase of AMAN group, unexpectedly, the patients had a significantly increased percentage of TNFR2 (P<0.01)and intracellular TNF-α(P< 0.01) expression on circulating monocytes after IVIg therapy. The level of TNFR2 on monocytes was increased dramatically (P< 0.05) before IVIg. After the immunomodulator intervention, the high level of TNFR2 was further up-regulated significantly(P< 0.01) compared with normal standard (healthy controls). Contrarily, the intracellular TNF-αpercentage of circulating monocytes was increased significantly (P< 0.01) only in the patients after IVIg treatment, but not pre-treatment. IVIg therapy down-regulated soluble Il-12p70 and TNFR1 in AIDP
     To further investigate the role of IL-12 and TNF-αin GBS and to determine the levels of IL-12 and TNF-α, as well as their receptors in sera, the enzyme linked immunosorbent assays (ELISA) of serous forms were performed in the GBS patients with AIDP, AMAN and healthy controls. The intravenous immunoglobulin therapy down-regulated the levels of soluble TNFR1 (P < 0.05 at T2) and IL-12p70 (P < 0.05 at T1, P < 0.01 at T2) into the normal standard (healthy controls). There were no significant results found in the AMAN group. Discussion
     Inflammatory cytokines plays an important role in initiating, enhancing and perpetuating pathogenic events in GBS. Our results showed that TNF-αand IL-12 molecules as well as their receptors are involved in the pathogenesis of GBS. Our results suggest TNF-αand IL-12p70 may play an inflammatory role in the pathogenesis of AIDP by increasing the expression of their receptor 1. In addition, TNF-αmay have an anti-inflammatory effect through a TNFR2-dependent mechanism in AMAN.
     TNF-αplays a complex and different role in AIDP and AMAN as it does in other autoimmune diseases. Because of its macrophage-stimulating capacity and inflammatory cell accumulation, TNF-αcan be classified as a Th1-type, pro-inflammatory cytokine that binds to its specific cell surface receptor, TNFR1. In patients with GBS it was found that TNF-αlevels correlate with clinical severity and serum concentration regression of TNF-αwas found correlated with the improvement of clinical features. However, recent studies have suggested anti-inflammatory and neuroprotective role of TNFR2, of which serum level assessment may serve as a prognostic factor of GBS. Similar to another study, our data demonstrated that the level of TNF-αand TNFR1 in AIDP was not increased in acute phase but in plateau phase, which was subsequently down-regulated by IVIg during plateau phase. On the other hand, TNF-αand TNFR2 were significantly up-regulated by the treatment of IVIg in AMAN during acute phase. Therefore, two conflicting mechanisms of TNF-αmay be involved in the pathogenesis of immunoglobulin therapy: a down-regulated TNFR1 dependent pathway in AIDP during plateau phase and an up-regulated TNFR2 dependent pathway in AMAN during acute phase.
     IL-12, or natural killer (NK) stimulatory factor, is a pleiotropic cytokine with Th1-promoting activity and acts as a proinflammatory stimulus, inducing consequent activation of NK cells and production of IFN-γin immune response. Administration of IL-12 exacerbated chronic EAN in Lewis rats and was associated with increased IFN-γproduction in the PNS. Paralleled with its inflammatory role, our data demonstrated that IL-12 and one of its receptors, IL-12Rβ1, were found to be increased in AIDP during acute phase, and then decreased during plateau phase after IVIg. The correlation analysis reconfirms the possible therapeutic mechanism in AIDP, i.e., that IVIg reduces the inflammatory activity of IL-12 by down-modulating the IL-12Rβ1 dependent pathway. Considering the Th1-promoting role of IL-12, our data indicated cellular immunity may be involved in pathogenesis of AIDP rather than in AMAN. There was no IL-12Rβ2 relative data were found significantly in our study; it deserves further investigation to delineate the role of it in GBS.
     Finally, although different explanations are involved, TNF-αand IL-12 molecules, as well as their receptors, participate in the pathogenesis of immunoglobulin therapy in GBS. Because inflammatory cytokines, IL-12 and TNF-αand their receptors may participate in AIDP, immune-modulating therapy may decrease their disease-promoting effect by reducing TNFR1 and IL-12Rβ1. Additionally, the anti-inflammatory role of TNF-αrealized through TNFR2 in AMAN is possibly a therapeutic mechanism in the IVIg treatment of AMAN.
     In summary, both TNF-αand IL-12 may play an inflammatory role in AIDP and AMAN. Moreover TNF-αmay also be an anti-inflammatory factor in AMAN by combining with TNFR2. The immunoglobulin therapy probably exerts its protective effect by down-modulating bioactivity of IL-12 through an IL-12Rβ1 dependent mechanism in AIDP and up-modulating the anti-inflammatory activity of TNF-αthrough a TNFR2 pathway in AMAN.
引文
[1] Hahn AF. Guillain-Barr′e syndrome. Lancet 1998; 352 (9128)
    [2] Cheng Q, Wang DS, Jiang GX, et al. Distinct pattern of age-specific incidence of Guillain-Barrésyndrome in Harbin, China. J Neurol 2002; 249: 25–32.
    [3] Hemachudha T, Griffin DE, Chen WW, et al. Immunologic of rabies vaccination-induced Guillain-Barre′syndrome. Neurology 1988; 38 (3): 375-8
    [4] Goetz: Textbook of Clinical Neurology, 3rd ed.
    [5] McKhann GM, Cornblath DR, Ho TW, Li CY, Bai AY, Wu HS.Clinical and electrophysiological aspects of acute paralytic disease of children and young adults in northern China. Lancet 1991; 338: 593–97.
    [6] Blaser MJ, Olivares A, Taylor DN, Cornblath DR, McKhann GM.Campylobacter serology in patients with Chinese paralytic syndrome. Lancet 1991; 338: 308.
    [7] Satoshi Kuwahara. Guillain-Barr′e syndrome Epidemiology, Pathophysiology and Management. J Drugs 2004:64 (6) :597-610
    [8] Winer JB, Hughes RA, Anderson MJ, et al. A prospective study of acute idiopathic neuropathy. II: antecedent events. J Neurol Neurosurg Psychiatry 1988; 51 (5): 613-8
    [9] Jacobs BC, Rothbarth PH, van der Meche FG, et al. The spectrum of antecedent infections in Guillain-Barr′e syndrome a case control study: Neurology 1998; 51 (4): 1110-5
    [10] Ogawara K, Kuwabara S, Mori M, et al. Axonal Guillain-Barre syndrome: relation to anti-ganglioside antibodies and Campylobacter jejuni infection in Japan. Ann Neurol 2000; 48 (4): 624-31
    [11] Hughes RA, Rees JH. Clinical and epidemiologic features of Guillain-Barre′syndrome. J Infect Dis 1997; 176 Suppl. 2:s92-8
    [12] Ho TW, Mishu B, Li CY, et al. Guillain-Barr′e syndrome in northern China:relationship to Campylobacter jejuni infection and anti-glycolipid antibodies. Brain 1995; 118 (Pt 3): 597-605
    [13] Hahn AF.Guillain-Barre syndrome: an evolving concept. Current Opinion in Neurology,1997; 10 (5) :363-365
    [14] Chiba A, Kusunoki S, Obata H, et al. Serum anti-GQ1b IgG antibody is associated with ophthalmoplegia in Miller Fisher syndrome and Guillain-Barr′e syndrome: clinical and immunohistochemical studies. Neurology 1993; 43 (10): 1991-7
    [15] Jacobs BC, Endtz HP, van der Meche FG, et al. Serum antiGQ1b antibodies recognize surface epitopes on Campylobacter jejuni from patients with Miller Fisher syndrome. Ann Neurol 1995; 37: 260-5
    [16] Kuroki S, Saida T, Nukina M, et al. Campylobacter jejuni strains from patients with Guillain-Barre′syndrome belong mostly to Penner serogroup 19 and contain beta-N-acetyl-glucosamine residues. Ann Neurol 1993; 33 (3): 243-7
    [17] Kuwabara S, Yuki N, Koga M, et al. IgG anti-GM1 antibody is associated with reversible conduction failure and axonal degeneration in Guillain-Barr′e syndrome. Ann Neurol 1998; 44 (12): 202-8
    [18] Nachamkin I. Epidemiology of Campylobacter jejuni infections in the United States and other industrial nations. In: Tauxe RV, Nachamkin I, Blaser MJ, et al., editors. Campylobacter jejuni: current status and future trends. Washington, DC: American Society for Microbiology, 1992: 9-19
    [19] Kuroki S, Saida T, Nukina M, et al. Campylobacter jejuni strains from patients with Guillain-Barre syndrome belong mostly to Penner serogroup 19 and contain p-Nacetylglucosamine residues. Ann Neurol 1993;33:243-247.
    [20] Sheikh KA, Nachamkin I, Ho TW, et al. Campylobacter jejuni lipopolysaccharides in Guillain-Barr′e syndrome: molecular mimicry and host susceptibility. Neurology 1998;51 (2): 371-8
    [21] Irie S, Saito T, Nakamura K, et al. Association of anti-GM2 antibodies in Guillain-Barre′syndrome with acute cytomegalovirus infection. J Neuroimmunol 1996; 68 (1-2): 19-26
    [22] Mori M, Kuwabara S, Miyake M, et al. Haemophilus influenzae infection and Guillain-Barre′syndrome. Brain 2000; 123 (Pt 10): 2171-8
    [23] Mori M, Kuwabara S, Miyake M, et al. Haemophilus influenzae has a GM1 ganglioside-like structure and elicits Guillain-Barre syndrome. Neurology 1999; 52 (6): 1282-4
    [24] Safranek TJ, Lawrence DN, Kurkland LT, et al. Reassessment of the association between Guillain-Barre′syndrome and receipt of swine influenza vaccine in 1976-1977: results of a two-state study. Expert Neurology Group. Am J Epidemiol 1991; 133: 952-5
    [25] Roscelli JD, Bass JW, Pang R. Guillain-Barre′syndrome and influenza vaccination in the US Army, 1980-1988. Am J Epidemiol 1991; 133 (9): 952-5
    [26] Haber P, DeStefano F, Angulo FJ, et al. Guillain-Barrésyndrome following influenza vaccination. JAMA 2004; 292: 2478–81.
    [27] Wijdicks EFM, Fletcher DD, Lawn ND. Influenzae vaccine and the risk of Guillain-Barr′e syndrome. Neurology 2000; 55 (3): 452-3
    [28] Pritchard R, Mukherjee R, Hughes RA. Risk of relapse of Guillain-Barr′e syndrome or chronic inflammatory demyelinating polyradiculoneuropathy following immunization. J Neurol Neurosurg Psychiatry 2002; 73: 348-9
    [29] Hadden RD, Cornblath DR, Hughes RA, et al. Electrophysiological classification of Guillain-Barre′syndrome: clinical associations and outcome: Plasma Exchange/Sandglobulin Guillain Barr′e Syndrome Trial Group. Ann Neurol 1998; 44 (5): 780-8
    [30] Ropper AH. Diseases of spinal cord, peripheral nerve, and muscle. In: Victor M, Ropper AH, editors. Principle of neuro-logy. New York: McGraw-Hill, 2001: 1380-7
    [31] Asbury AK, Arnason BG, Adams RD. The inflammatory lesion in idiopathic polyneuritis. Its role in pathogenesis. Medicine 1969; 48:173–215.
    [32] Prineas JW. Pathology of the Guillain-Barre′syndrome. Ann Neurol 1981; 9 Suppl.: 6-19
    [33] Kieseier BC, Kiefer R, Gold R, Hemmer B, Willison HJ,Hartung HP. Advances in understanding and treatment of immune-mediated disorders of the peripheral nervous system. Muscle Nerve 2004; 30: 131–56.
    [34] Hafer-Macko CE, Sheikh KA, Li CY, et al. Immune attack on the Schwann cell surface in acute inflammatory demyelinating
    [35] polyneuropathy. Ann Neurol 1996; 39: 627–37.
    [36] Kuwabara S, Nakata M, Sung JY, et al. Hyperreflexia in axonal Guillain-Barr′e syndrome subsequent to Campylobacter jejuni enteritis. J Neurol Sci 2002; 199 (1-2): 89-92
    [37] Griffin JW, Li CY, Macko C, et al. Early nodal changes in the acute motor axonal neuropathy pattern of the Guillain-Barrésyndrome. J Neurocytol 1996; 25: 33–51.
    [38] Griffin JW, Li CY, Ho TW, et al. Pathology of the motor sensory axonal Guillain-Barr′e syndrome. Ann Neurol 1996; 39 (1): 17-28
    [39] Yuki N, Taki T, Inagaki F, et al. A bacterium lipopolysaccharide that elicits Guillain-Barr′e syndrome has a GM1 ganglioside-like structure. J Exp Med 1993; 178 (5): 1771-5
    [40] 39 Fross RD, Daube J. Neuropathy in the Miller-Fisher syndrome: clinical and electrophysiologic findings. Neurology 1987; 37:1493–98
    [41] Paradiso G, Tripoli J, Galicchio S, Fejerman N. Epidemiological, clinical, andelectrodiagnostic findings in childhood Guillain-Barrésyndrome: a reappraisal. Ann Neurol 1999; 46: 701–07.
    [42] The Guillain-Barr′e Syndrome Study Group. Plasmapheresis and Guillain-Barr′e syndrome. Neurology 1985; 35: 1096-2004
    [43] Richard A. C. Hughes, Anthony V. Swan, et al. Immunotherapy for Guillain-Barre¤syndrome: a systematic review. Brain (2007), 130, 2245-2257
    [44] Dalakas MC. Mechanisms of action of IVIg and therapeutic considerations in the treatment of acute and chronic demyelinating neuropathies. Neurology 2002; 59 (12 Suppl. 6): S13-21
    [45] Hughes RA, van der Meche FG. Corticosteroids for Guillain-Barre′syndrome. Cochrane Database of Systematic Reviews. Available in The Cochrane Library [database on disk and CD ROM]. Updated quarterly. The Cochrane Collaboration; issue 3. Oxford: Update Software, 2003
    [46] Van Koningsveld R, van der Meche FG, Schmitz PI, et al.Combined therapy of intravenous immunoglobulin and methylprednisolone in patients with Guillain-Barre′syndrome: the results of a multicentre double blind placebo controlled clinical trial. J Peripher Nerv Syst 2001; 6: 186-187
    [1]黄克维,吴丽娟,临床神经病理学,北京:人民军医出版社,1999.1,238-239
    [2] McKhann GM, Cornblath DR, Ho Tw. Clinical and electrophysiological aspects of acute paralytic disease of children and young adults in northern China. Lancet 1991 Sep 7; 338(8767): 593–597.
    [3] Blaser MJ, Olivares A, Taylor DN.Campylobacter serology in patients with Chinese paralytic syndrome. Lancet 1991 Aug 3; 338(8762): 308.
    [4] Shin I-S J, Baer AN, Kwon HJ, Papadopoulos EJ, Siegel JN. Guillain–Barre and Miller Fisher syndromes occurring with tumor necrosis factor antagonist therapy. Arthritis Rheum 2006;54:1429–1434.
    [5] Asbury AK, Arnason BGW, Adams RD. The inflammatory lesion in idiopathic neuritis its role in pathogenesis. Medicine 1969;48:173-215
    [6] Anna Makowska, Jane Pritchard, Lara Sanvito, et al. Immune responses to myelin proteins in Guillain-Barre syndrome. J Neurol Neurosurg Psychiatry. 2008 Jun;79(6):664-71
    [7] Willison HJ, Yuki N. Peripheral neuropathies and anti-glycolipid antibodies. Brain.2002;125:2591-2625
    [8] Trinchieri G, Scott P. Interleukin-12: basic principles and clinical applications. Curr Top Microbiol Immunol 1999;238:57–78.
    [9] Ha SJ, et al. A novel function of IL-12p40 as a chemotactic molecule for macrophages. J Immunol 1999;163(5):2902–8.
    [10] Esche C, Shurin MR, Lotze MT. IL-12 Receptor. In: Oppenheim JJ, Feldmann M, editors. Cytokine reference. San Diego: Academic Press; 2000.
    [11] Presky DH, et al. A functional interleukin-12 receptor complex is composed of twoβ-type cytokine receptor subunits. Proc Natl Acad Sci USA 1996;93(24):14002–7.
    [12] Grohmann U, et al. IL-12 acts directly on DC to promote nuclear localization of NF-κB and primes DC for IL-12 production. Immunity 1998;9(3):315–23.
    [13] Rogge L, et al. Antibodies to the IL-12 receptor-β2 chain mark human Th1 but not Th2 cells in vitro and in vivo. J Immunol 1999;162(7):3926–32.
    [14] Afkarian M, et al. T-bet is a STAT1-induced regulator of IL-12 receptor expression in naive CD4+ T cells. Nat Immunol; 2002.
    [15] Rogge L, et al. Selective expression of an interleukin-12 receptor component by humanT helper 1 cells. J Exp Med 1997;185(5): 825–31.
    [16] Sinigaglia F, et al. Regulation of the IL-12–IL-12R axis: a critical step in T-helper cell differentiation and effector function. Immunol Rev 1999;170:65–72.
    [17] Dong C, Flavell RA. Th1 and Th2 cells. Curr Opin Hematol 2001;8(1):47–51.
    [18] O’Shea JJ, Gadina M, Schreiber RD. Cytokine signaling in 2002: new surprises in the JAK–STAT pathway. Cell 2002;109(Suppl): S121–131.
    [19] O’Shea JJ, Paul WE. Regulation of T(H)1 differentiation controlling the controllers. Nat Immunol 2002;3(6):506–8.
    [20] Farrar JD, Asnagli H, Murphy KM. T helper subset development: roles of instruction, selection, and transcription. J Clin Invest 2002;109(4):431–5.
    [21] Lawless VA, et al. STAT4 regulates multiple components of IFN-γ-inducing signaling pathways. J Immunol 2000;165(12): 6803–8.
    [22] Fukao T, et al. Inducible expression of STAT4 in dendritic cells and macrophages and its critical role in innate and adaptive immune responses. J Immunol 2001;166(7):4446–55.
    [23] Frucht DM, et al. IFN-γproduction by antigen-presenting cells: mechanisms emerge. Trends Immunol 2001;22(10):556–60.
    [24] Du C, Cooper JC, Klaus SJ and Sriram S. Amelioration of CR-EAE with lisofylline: effects on mRNA levels of IL-12 and IFN-gamma in the CNS. J Neuroimmunol, 2000, 110:13-19.
    [25] Segal BM, Dwyer BK and Shevach EM. An interleukin (IL)-10/IL-12 immunoregulatory circuit controls susceptibility to autoimmune disease. J Exp Med , 1998; 187:537-546.
    [26] Constantinescu CS, Hilliard B, Ventura E, Wysocka M, Showe L, Lavi E, Fujioka T, Scott P, Trinchieri G, Rostami A. Modulation of susceptibility and resistance to an autoimmune model of multiple sclerosis in prototypically susceptible and resistant strains by neutralization of interleukin-12 and interleukin-4, respectively. Clin Immunol , 2001; 98:23-30.
    [27] Debra MB, Anne MM, Ravinder NM, Fionula MB and Marc F. Anti-IL-12 and anti-TNF antibodies synergistically suppress the progression of murine collageninduced arthritis. Eur J Immunol, 1999; 29:2205-2212.
    [28] Zhu J, Bai XF, Mix E et al. Cytokine dichotomy in the peripheral nervous tissues influence the outcome of experimental allergic neuritis: dynamics of mRNA expressionfor IL-1b, IL-6, IL-10, IL-12, TNF-a and TNF-b and cytolysin. Clin Immunol Immunopathol 1997;84:85–94.
    [29] Zhu J, Mix E, Link H. Cytokine production and the pathogenesis of experimental autoimmune neuritis and Guillain–Barre′syndrome. J Neuroimmunol 1998;84:40–52.
    [30] Zhu J, Bai XF, Mix E et al. Experimental allergic neuritis: cytolysin mRNA expression is upregulated in lymph node cells during convalescence. J Neuroimmunol 1997;78:108–16.
    [31] Trinchieri G.. Interleukin-12: a proinflammatory cytokine with immunoregulatory functions that bridge innate resistance and antigenspecific adaptive immunity. Annu Rev Immunol 1995;13:251–76.
    [32] Morris SC, Madden KB, Adamovicz JJ et al. Effects of IL-12 on in vivo cytokine gene expression and Ig isotype selection. J Immunol 1994;152:1047–56.
    [33] S. H. Pelidou, G. Deretzi, L. P. Zou, et al. Inflammation and severe demyelination in the peripheral nervous system induced by the intraneural injection of recombinant mouse interleukin-12. Scand. J. Immunol. 1999, 50, 39-44
    [34] Zhu J, Bai XF, Hedlund G, Bj?rk J, Bakhiet M, van der Meide P, Link H. Linomide suppresses experimental allergic neuritis in Lewis rats by inhibiting myelin antigen- reactive T and B cells responses. Clin Exp Immunol (1999)1: 56-63.
    [35] Zhu Y, Ljunggren HG, Mix E, Li HL, van der Meide P, Elhassan AM, Winblad B, Zhu J (2001) CD28/B7 costimulation: a critical role for initiation and development of experimental autoimmune neuritis in C57BL/6 mice. J Neuroimmunol 114:114-121.
    [36] Lei Bao, J.Urban Lindgren, Peter van der Meile, et al. The critical role of IL-12p40 in initiating, enhancing, and perpetuating pathogenic events in murine experimental autoimmune neuritis. Brain Pathol 2002;12:420-429
    [37]刘建新,郑昌学,现代免疫学-免疫的细胞和分子基础,北京:清华大学出版社,2002.3,308-309.
    [38] Farrah T, Smith C. Emerging cytokine family.Nature. 1992 Jul 2;358(6381):26
    [39] Smith C, Gruss H, Davis T, et al.CD30 antigen, a marker for Hodgkin's lymphoma, is a receptor whose ligand defines an emerging family of cytokines with homology to TNF. Cell. 1993 Jul 2;73(7):1349-60.
    [40] Dan Aderka. The potential biological and clinical significance of the soluble tumor necrosis factor receptors.Cytokine & Growth Factor Review. 1996,7(3):231-240
    [41] Robinson WH, Genovese MC, Moreland LW. Demyelinating and neurologic eventsreported in association with tumor necrosis factor-αantagonism. Arthritis Rheum 2001;44:1977– 1983.
    [42] Kollias G., Kontoyiannis D. Role of TNF/TNFR in autoimmunity: specific TNF receptor blockade may be advantageous to anti-TNF treatments. Cytokine & Growth Factor Reviews 2002; 13: 315–321.
    [43] Myers RR, Campana WM, Shubayev VI. The role of neuroinflammation in neuropathic pain: mechanisms and therapeutic targets. Drug Discovery Today 2006; 11: 8–20.
    [44] Radhakrishnan VV, Sumi MG, Reuben S, Mathai A, Nair MD. Serum tumour necrosis factor-alpha and soluble tumour necrosis factor receptors levels in patients with Guillain-Barre syndrome. Acta Neurologica Scandinavica 2004; 109: 71–74.
    [45] Cralg A, Smith, Terry Farrah, et al.The TNF receptor superfamily of cellular and viral proteins:activation, costimulation, and death. Cell.1994 March 25;76:959-962
    [46] Cope A, Ettinger R, McDevitt H. The role of TNF-αand related cytokines in the development and function of the autoreactive T-cell repertoire. Res Immunol 1997;148:307– 312.
    [47] Kollias G, Douni E, Kassiotis G, Kontoyiannis D. The role of tumor necrosis factor and receptors in models of multi-organ inflammation, rheumatoid arthritis, multiple sclerosis, and inflammatory bowel disease. Ann Rheum Dis 1999;58(suppl 1):32–39.
    [48] Cope AP, Liblau RS, Yang XD, Congia M, Laudanna C, Schreiber RD. Chronic tumor necrosis factor alter T cell responses by attenuating T cell receptor signaling. J Exp Med 1997;185: 1573–1584.
    [49] Enbrel (etanercept). Current prescribing information. In: Physicians’desk reference. Montvale, NJ: Thompson; 2007. p 584–591.
    [50] HUMIRA (adalimumab). Current prescribing information. In: Physicians’desk reference. Montvale, NJ: Thompson; 2007. p 466–471.
    [51] Moser B, Wolf M, Walz A.Chemokines: multiple levels of leukocyte migration control. Trends Immunol 2004; 25:75– 84.
    [52] De Groote D, Grau GE, Dehart I, Franchimont P. Sta-bilization of functional tumor necrosis factor-a by its sol-uble receptors. Eur Cytokine Netw 1993, 4, 359-362.
    [53] Aderka D, Engelmann H, Maor Y, Brakebusch C, Wal-lach D. Stabilization of the bioactivity of tumor necrosis factor by its soluble receptors. JExp Med 1992, 175, 323-329
    [54] Porteu F, Brockhaus M, Wallach D, Englemann H, Nathan CF. Human neutrophilelastase releases a ligand-binding fragment from the 75-kDa tumor necrosis factor (TNF) receptor. J Biol Chem 1991, 266, 18846-18853.
    [55] Crowe PD, Walter BN, Mohler KM, Otten-Evans C, Black RA, Ware CF. A rnetalloprotease inhibitor blocks shedding of the 80-kDa TNF receptor and TNF processing in T lymphocytes. J Exp Med 1995, 181, 1205-1210
    [56] Aderka D, Englemann H, Hornik V, Skornick Y, Levo Y, Wallach D, Kushtai G. Increased serum levels of soluble receptors for tumor necrosis factor in cancer patients, Cancer Res 1991, 51, 5602-5607.
    [57] Aderka D, Englemann H, Wallach D. Soluble tumor necrosis factor receptors in health and disease. In Friers W, Buurman WA, eds. Tumor Necrosis Factor: Molecular and Cellular Biology andC/inical Re/erance. Karger, Basel, 1993, 191-198.
    [58] Kieseier BC, Kiefer R, Gold R, Hemmer B, Willison HJ, Hartung H-P. Advances in understanding and treatment of immune- mediated disorders of the peripheral nervous system. Muscle Nerve 2004;30:131–156.
    [59] Kieseier BC, Krivacic K, Jung S, Pischel H, Toyka KV, Ransohoff RM, et al. Sequential expression of chemokines in experimental autoimmune neuritis. J Neuroimmunol, 2000;110: 121–129.
    [60] Hartung H-P. Immune mediated demyelination. Ann Neurol 1993;33:563–567.
    [61] Vasalli P. The pathophysiology of tumor necrosis factors. Annu Rev Immunol 1992;10:411– 452.
    [62] Redford EJ, Hall SM, Smith KJ. Vascular changes and demyelination induced by the intraneural injection of tumor necrosis factor. Brain 1995;118:869–878.
    [63] Tsang RS, Valdivieso-Garcia A. Pathogenesis of Guillain– Barre′syndrome. Expert Rev Anti Infect Ther 2003;1:597– 608.
    [64] Hall SM, Redford EJ, Smith KJ. Tumor necrosis factor-a has few morphological effects within the dorsal columns of the spinal cord, in contrast to its effects in the peripheral nervous system. J Neuroimmunol 2000;106:130–136.
    [65] Selmaj KW, Raine CS. Tumor necrosis factor mediates myelin and oligodendrocyte damage in vitro. Ann Neurol 1988;23: 339–346.
    [66] Bronan CF, Litwak MS, Schroeder CE, Selmaj K, Raine CS, Arezzo JC. Preliminary studies of cytokine-induced functional effects of the visual pathways in the rabbit. J Neuroimmunol 1989;25:227–239.
    [67] Said G, Hontebeyrie-Joskowicz M. Nerve lesions induced by macrophage activation.Res Immunol 1992;143:589–599.
    [68] M. Empl, S. Renaud, B.Erne. et al. TNF-alpha expression in painful and nonpainful neuropathies. Neurology 2001;56;1371-1377
    [69] Skoff AM, Lisak RP, Bealmear B, et al.TNF-alpha and TGF-beta act synergistically to kill Schwann cells, J Neurosci Res. 1998 Sep 5;53(6):747-56
    [70] Stoll G, Jung S, Jander S, van der Meide P, Hartung HP. Tumor necrosis factor-alpha in immune-mediated demyelination and Wallerian degeneration of the rat peripheral nervous system. J Neuroimmunol 1993;45:175–182.
    [71] Li-Ping Zou, Georgia Deretzi, Sigliti-Henrietta Pelidou, Rolipram suppresses experimental autoimmune neuritis and prevents relapses in Lewis rats. Neuropharmacology, 2000;39:324–333
    [72] Weishaupt A, Bruck W, Hartung T, Toyka KV, Gold R. Schwann cell apoptosis in experimental autoimmune neuritis of the Lewis rat and the functional role of tumor necrosis factor-a. Neurosci Lett 2001;306:77– 80.
    [73] Lei Bao,J. Urban Lindgren, Yu Zhu, et al. Exogenous soluble tumor necrosis factor receptor type I ameliorates murine experimental autoimmune neuritis. Neurobiology of Disease , 2003;12:73–81
    [74] Sorkin LS, Doom CM. Epineural application of TNF elicits an acute mechanical hyperalgesia in the awake rat. J Peripher Nerv Syst 2000;5:96–100.
    [75] Sommer C, Schafers M, Marziniak M, Toyka KV. Etanercept reduces hyperalgesia in experimental painful neuropathy. J Peripher Nerv Syst 2001;6:67–72.
    [76] Misawa S, Kuwabara S, Mori M, Kawagushi N, Yoshiyama Y, Hattori T. Serum levels of tumor necrosis factor-αin chronic inflammatory polyneuropathy. Neurology 2001;56:666–669.
    [77] Sharief MK, McLean B, Thompson EJ. Elevated serum levels of tumor necrosis factor-αin Guillain–Barre′syndrome. Ann Neurol 1993;33:591–596.
    [78] Terenghi F, Allaria S, Nobile-Orazio E. Circulating levels of cytokines and their modulation by intravenous immunoglobulin in multifocal motor neuropathy. J Periph Nerv Syst 2006; 11:67–71.
    [79] Zhu J, Mix E, Link H. Cytokine production and the pathogenesis of experimental autoimmune neuritis and Guillain–Barre′syndrome. J Neuroimmunol 1998;84:40–52.
    [80] Cre′ange A, Be′lec L, Clair B, Raphael JC, Gherardi RK. Circulating tumor necrosis factor (TNF)-a and soluble TNF-a receptors in patients with Guillain–Barre′syndrome.J Neuroimmunol 1996;68:95–99.
    [81] Reuben S, Sumi MG, Mathai A, et al. Intravenous immunoglobulin reduces serum tumor necrosis factor a in patients with Guillain-Barre syndrome. Neurol India, 2003,51(4):487-489
    [82] .Exley A, Smith N, Winer J. Tumor necrosis factor-a and other cytokines in Guillain-Barre syndrome. Ann Neurol 1993;33:591-596
    [83] Sharief MK, Ingram DA, Swash M. Circulating tumor necrosis factor-a correlates with electrodiagnostic abnormalities in Guillain–Barre′syndrome. Ann Neurol 1997;42:68–73.
    [84] Sharief MK, Ingram DA, Swash DA, Thompson EJ. IV immunoglobulin reduces circulating proinflammatory cytokines in Guillain–Barre′syndrome. Neurology 1999;52:1833–1838.
    [85] Mathey EK, Pollard JD, Armati PJ. TNF alpha, IFN gamma, and IL-2 mRNA expression in CIDP sural nerve biopsies. J Neurol Sci 1999;163:47–52.
    [86] Alldred A. Etanercept in rheumatoid arthritis. Expert Opin Pharmacother 2001;1:1137–1148.
    [87] Feagan BG, Enns R, Fedorak RN, Panaccione R, Pare P, Steinhart AH, et al. Infliximab for the treatment of Crohn’s disease: efficacy, safety and pharmacoeconomics. Can J Clin Pharmacol 2001;8:188–198.
    [88] Hamilton K, Clair EW. Tumor necrosis factor-alpha blockade: a new era for the effectivemanagement of rheumatoid arthritis. Expert Opin Pharmacother 2000;1:1041–1052.
    [89] In-Sook J. Shin, Alan N. Baer, Hyon J. Kwon, et al. Guillain-Barre and Miller Fisher syndromes occurring with tumor necrosis factor a antagonist therapy. Arthritis Rheumatism, 2006 May 54(5):1429-1434
    [90] Niveditha Mohan, Evelyne T. Edwards, et al. Demydlination occurring During Anti-tumor necrosis factor a therapy for inflammatory arthritidies. Arthritis Rheumatism, 2001 Dec
    [91] Remicade (infliximab). Current prescribing information. In: Physicians’desk reference. Montvale, NJ: Thompson;2007.p 971–979.44(12):pp2862-2869
    [92] Kassiotis G, Kollias G. Uncoupling the proinflammatory from the immunosuppressive properties of tumor necrosis factor (TNF) at the p55 TNF receptor level: implications for pathogenesis and therapy of autoimmune demyelination. J Exp Med 2001;193:427–434.
    [93] Liu J, Marino MW, Wong G. TNF is a potent anti-inflammatory cytokine in autoimmune-mediated demyelination. Nat Med 1998;4:78–83.6
    [94] Lu MO, Duan RS, Quezada HC.Aggravation of experimental autoimmune neuritis in TNF-alpha receptor 1 deficient mice. J Neuroimmunol. 2007 May;186(1-2):19-26
    [1] Hadden RD, Cornblath DR, Hughes RA, et al. Electrophysiological classification of Guillain-Barre′syndrome: clinical associations and outcome: Plasma Exchange/Sandglobulin Guillain Barr′e Syndrome Trial Group. Ann Neurol 1998; 44 (5): 780-788
    [2] Prineas JW. Pathology of the Guillain-Barre′syndrome. Ann Neurol 1981; 9 Suppl.: 6-19
    [3] Asbury AK, Arnason BG, Adams RD. The inflammatory lesion in idiopathic polyneuritis. Its role in pathogenesis. Medicine 1969; 48:173–215.
    [4] Kieseier BC, Kiefer R, Gold R, Hemmer B, Willison HJ,Hartung HP. Advances in understanding and treatment of immune-mediated disorders of the peripheral nervous system. Muscle Nerve 2004; 30: 131–156.
    [5] Hafer-Macko CE, Sheikh KA, Li CY, et al. Immune attack on the Schwann cell surface in acute inflammatory demyelinating polyneuropathy. Ann Neurol 1996; 39: 627–637.
    [6] Ho TW, Mishu B, Li CY, et al. Guillain-Barre syndrome in northern China: relationship to Campylobacter jejuni infection and anti-glycolipid antibodies. Brain 1995; 118 (Pt 3): 597-605
    [7] Kuwabara S, Nakata M, Sung JY, et al. Hyperreflexia in axonal Guillain-Barre syndrome subsequent to Campylobacter jejuni enteritis. J Neurol Sci 2002; 199 (1-2): 89-92
    [8] Ho, T.W, Li, C.Y,Cornblath, D.R, et al.Patterns of recovery in the Guillain-Barre syndrome. Neurology, 1997; 48(3):695-700
    [9] Ogawara K, Kuwabara S, Mori M, et al. Axonal Guillain-Barre syndrome: relation to anti-ganglioside antibodies and Campylobacter jejuni infection in Japan. Ann Neurol 2000; 48 (4): 624-631
    [10] Kuwabara S, Yuki N, Koga M, et al. IgG anti-GM1 antibody is associated with reversible conduction failure and axonal degeneration in Guillain-Barre syndrome. Ann Neurol 1998; 44 (12): 202-208
    [11] Griffin JW, Li CY, Macko C, et al. Early nodal changes in the acute motor axonal neuropathy pattern of the Guillain-Barrésyndrome. J Neurocytol 1996; 25: 33–51.
    [12] O’Shea JJ, Paul WE. Regulation of T(H)1 differentiation controlling the controllers. Nat Immunol 2002;3(6):506–508.-
    [13]刘建新,郑昌学,现代免疫学-免疫的细胞和分子基础,北京:清华大学出版社,2002.3,308-309.
    [14] Dong C, Flavell RA. Th1 and Th2 cells. Curr Opin Hematol 2001;8(1):47–51.
    [15] O’Shea JJ, Gadina M, Schreiber RD. Cytokine signaling in 2002: new surprises in the JAK–STAT pathway. Cell 2002;109(Suppl): S121–131.
    [16] Segal BM, Dwyer BK and Shevach EM. An interleukin (IL)-10/IL-12 immunoregulatory circuit controls susceptibility to autoimmune disease. J Exp Med , 1998; 187:537-546.
    [17] Li-Ping Zou, Georgia Deretzi, Sigliti-Henrietta Pelidou, Rolipram suppresses experimental autoimmune neuritis and prevents relapses in Lewis rats. Neuropharmacology, 2000;39:324–333
    [18] Weishaupt A, Bruck W, Hartung T, Toyka KV, Gold R. Schwann cell apoptosis in experimental autoimmune neuritis of the Lewis rat and the functional role of tumor necrosis factor-a. Neurosci Lett 2001;306:77– 80.
    [19] Misawa S, Kuwabara S, Mori M, Kawagushi N, Yoshiyama Y, Hattori T. Serum levels of tumor necrosis factor-αin chronic inflammatory polyneuropathy. Neurology 2001;56:666–669.
    [20] Terenghi F, Allaria S, Nobile-Orazio E. Circulating levels of cytokines and their modulation by intravenous immunoglobulin in multifocal motor neuropathy. J Periph Nerv Syst 2006; 11:67–71.
    [21] Lu MO, Duan RS, Quezada HC.Aggravation of experimental autoimmune neuritis in TNF-alpha receptor 1 deficient mice. J Neuroimmunol. 2007 May;186(1-2):19-26
    [22] In-Sook J. Shin, Alan N. Baer, Hyon J. Kwon, et al. Guillain-Barre and Miller Fisher syndromes occurring with tumor necrosis factor a antagonist therapy. Arthritis Rheumatism, 2006 May 54(5):1429-1434
    [23] Zhu J, Bai XF, Mix E et al. Experimental allergic neuritis: cytolysin mRNA expression is upregulated in lymph node cells during convalescence. J Neuroimmunol 1997;78:108–116.
    [24] Morris SC, Madden KB, Adamovicz JJ et al. Effects of IL-12 on in vivo cytokine gene expression and Ig isotype selection. J Immunol 1994;152:1047–1056.
    [25] S. H. Pelidou, G. Deretzi, L. P. Zou, et al. Inflammation and severe demyelination in the peripheral nervous system induced by the intraneural injection of recombinant mouse interleukin-12. Scand. J. Immunol. 1999, 50, 39-44
    [26] Zhu Y, Ljunggren HG, Mix E, et al.CD28/B7 costimulation: a critical role for initiation and development of experimental autoimmune neuritis in C57BL/6 mice. JNeuroimmunol 2001;114:114-121.
    [27] Lei Bao, J.Urban Lindgren, Peter van der Meile, et al. The critical role of IL-12p40 in initiating, enhancing, and perpetuating pathogenic events in murine experimental autoimmune neuritis. Brain Pathol 2002;12:420-429
    [28] Kollias G., Kontoyiannis D. Role of TNF/TNFR in autoimmunity: specific TNF receptor blockade may be advantageous to anti-TNF treatments. Cytokine & Growth Factor Reviews 2002; 13: 315–321.
    [29] Misawa S, Kuwabara S, Mori M, et al. Serum levels of tumor necrosis factor-alpha in chronic inflammatory demyelinating polyneuropathy. Neurology 2001; 56: 666–669.
    [30] V.V. Radhakrishnan, M.G. Sumi, S. Reuben. Circulating tumour necrosis factor alpha & soluble TNF receptors in patients with Guillain-Barre syndrome. Indian J Med Res 2003;117:pp 216-220
    [31] Exley AR, Smith N, Winer JB. Tumor necrosis factoralpha and other cytokines in Guillain-Barre syndrome. Journal of Neurology, Neurosurgery, and Psychiatry 1994; 57: 1118–1120.
    [32] Durand MC, Lofaso F, Lefaucheur JP, et al. Electrophysiology to predict mechanical ventilation in Guillain- Barre syndrome. European Journal of Neurology 2003; 10: 39–44.
    [33] Sharief MK, Ingram DA, Swash M, et al. I.v.immunoglobulin reduces circulating proinflammatory cytokines in Guillain-Barre syndrome. Neurology 1999; 52: 1833–1838.
    [34] Myers RR, Campana WM, Shubayev VI. The role of neuroinflammation in neuropathic pain: mechanisms and therapeutic targets. Drug Discovery Today 2006; 11:8-20.
    [35] Radhakrishnan VV, Sumi MG, Reuben S, Mathai A, Nair MD. Serum tumour necrosis factor-alpha and soluble tumour necrosis factor receptors levels in patients with Guillain-Barre syndrome. Acta Neurologica Scandinavica 2004; 109: 71–74.
    [36] Press R, Ozenci V, Kouwenhoven M, Link H. Non-T(H)1 cytokines are augmented systematically early in Guillain-Barre syndrome. Neurology 2002; 58: 476–478.
    [37] Dalakas MC. Mechanisms of action of IVIg and therapeutic considerations in the treatment of acute and chronic demyelinating neuropathies. Neurology 2002; 59 (12 Suppl. 6): S13-21

© 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