Campylobacter gene polymorphism as a determinant of clinical features of Guillain–Barré syndrome M. Koga, MD, PhD; M. Takahashi, DVM, PhD; M. Masuda, MD, PhD; K. Hirata, MD, PhD; and N. Yuki, MD, PhD Abstract—Background: Ganglioside epitopes on Campylobacter jejuni are hypothesized as the key to the development and characterization of Guillain–Barré syndrome (GBS), but a comprehensive theory has yet to be established. A C jejuni gene, cst-II, involved in the biosynthesis of ganglioside-like lipo-oligosaccharide, shows a polymorphism (Asn/Thr51) that affects ganglioside epitopes. Objective: To examine the hypothesis that this polymorphism determines autoantibody reactivity, and thereby neurologic presentations in GBS. Methods: C jejuni isolates were collected from 105 GBS (including its variants) and 65 uncomplicated enteritis patients. The authors examined the frequency of cst-II and polymorphism (Asn/Thr51) in connection with the bacterial ganglioside epitopes, autoantibody reactivities against GM1, GD1a, and GQ1b, and patients’ neurologic findings. Results: Neuropathic strains more frequently had cst-II, in particular cst-II (Thr51), than did enteritic ones (85% vs 52%; p ⬍ 0.001). Strains with cst-II (Asn51) regularly expressed the GQ1b epitope (83%), whereas those with cst-II (Thr51) had the GM1 (92%) and GD1a (91%) epitopes. The presence of these bacterial epitopes in neuropathy patients corresponded to autoantibody reactivity. Patients infected with C jejuni (Asn51) more often were positive for anti-GQ1b IgG (56% vs 8%; p ⬍ 0.001) and had ophthalmoparesis (64% vs 13%; p ⬍ 0.001) and ataxia (42% vs 11%; p ⫽ 0.001). Patients who had C jejuni (Thr51) more frequently were positive for anti-GM1 (88% vs 35%; p ⬍ 0.001) and anti-GD1a IgG (52% vs 24%; p ⫽ 0.006) and had limb weakness (98% vs 71%; p ⬍ 0.001). Conclusions: The genetic polymorphism of C jejuni determines autoantibody reactivity as well as the clinical presentation of Guillain–Barré syndrome (GBS), possibly through modification of the host-mimicking molecule. The GBS paradigm is the first to explain the detailed pathogenesis of a postinfectious, autoimmune-mediated, molecular mimicry-triggering disorder. NEUROLOGY 2005;65:1376–1381 Guillain–Barré syndrome (GBS) is characterized by limb weakness and loss of tendon reflexes, but has a variety of other neurologic presentations.1 Anti-GM1 and anti-GD1a IgG antibodies are associated with axonal GBS,2 whereas anti-GQ1b IgG antibody is specific to patients with Fisher syndrome (FS) or GBS with ophthalmoplegia.3 Campylobacter jejuni is the most frequent antecedent infectious agent in GBS.4 The critical factor that causes the development of neuropathy after C jejuni infection is unAdditional material related to this article can be found on the Neurology Web site. Go to www.neurology.org and scroll down the Table of Contents for the November 8 issue to find the title link for this article. known, but the bacterial lipo-oligosaccharide (LOS) is a candidate because its terminal sugar regions mimic the sugar residues of the gangliosides GM1, GD1a, and GQ1b.5-7 The development of axonal GBS model after inoculation of rabbits with the GM1-like LOS has provided conclusive evidence for the hypothesis that the ganglioside-mimicry of C jejuni LOS is a cause of GBS.8 However, determinant factor of anti-ganglioside antibody specificity and neurologic presentation in GBS remains unclear. Determination of the complete genome sequence of C jejuni NCTC 11168 showed that many LOS biosynthesis genes are encoded in a large cluster.9 A subsequent study identified the genes involved in the Editorial, see page 1350 This article was previously published in electronic format as an Expedited E-Pub on September 14, 2005, at www.neurology.org. From the Departments of Neurology (Drs. Koga, Hirata, and Yuki) and Microbiology (Dr. Masuda), Dokkyo University School of Medicine, Tochigi; and Department of Microbiology (Dr. Takahashi), Tokyo Metropolitan Institute of Public Health, Japan. Supported in part by grants from the Ichiro Kanehara Foundation to M.K.; the Kanae Foundation for Life & Socio-Medical Science to M.K.; the Japan Intractable Diseases Research Foundation to M.K.; Mizutani Foundation for Glycoscience to N.Y.; a grant for Scientific Research (B) (KAKENHI 14370210 to N.Y.) from the Ministry of Education, Culture, Sports, Science and Technology of Japan; a Research Grant for Neuroimmunological Diseases to N.Y. from the Ministry of Health, Labour and Welfare of Japan; a Health Sciences Research Grant (Research on Psychiatric and Neurological Diseases and Mental Health) to N.Y. from the Ministry of Health, Labour, and Welfare of Japan; and a grant from the Human Frontier Science Program (RGP0038/2003-C to N.Y.). Disclosure: The authors report no conflicts of interest. Received April 4, 2005. Accepted in final form June 20, 2005. Address correspondence and reprint requests to Dr. M. Koga, Department of Neurology, Dokkyo University School of Medicine, Kitakobayashi 880, Mibu, Shimotsuga, Tochigi 321-0293, Japan; e-mail: kogamrk@dokkyomed.ac.jp 1376 Copyright © 2005 by AAN Enterprises, Inc. transfer of galactose, N-acetylgalactosamine, and sialic acid to the LOS outer core.10 Because ganglioside classification is based on the sialylation type (see figure E-1 on the Neurology Web site at www. neurology.org),11 sialyltransferase-encoding genes may be associated with the variation in the ganglioside epitope on LOS. It was reported that cst-II, a gene encoding sialyltransferase,10,12 was present in all eight strains with GQ1b-like LOS and proposed that its presence is associated with various ganglioside-like LOSs, although cst-II frequency did not differ between the GBS/FS and uncomplicated enteritis strains studied.13 In contrast, based on tests of 28 GBS isolates, it was reported that the cst-II gene is more often present in GBS isolates than in enteritis isolates.14 Whether the presence of this gene is a risk factor for developing neuropathy after C jejuni enteritis has yet to be proved. The ganglioside-like structure of the C jejuni LOS is, in part, determined by the multiple mechanisms the bacterium uses to turn on or off a gene or to modulate the substrate specificities of its glycosyltransferases, as well as by different gene contents.15 Interestingly, variation in the nucleotide sequence of cst-II might affect enzymatic activity; Cst-II (Thr51) has only ␣-2,3-sialyltransferase activity (monofunctional), whereas Cst-II (Asn51) has both ␣-2,3- and ␣-2,8-sialyltransferase activities (bifunctional).15 Because both ␣-2,3- and ␣-2,8-sialyltransferase activities are required for the biosynthesis of GQ1b mimics such as GT1a- or GD1c-like LOS,6,7 cst-II polymorphism is assumed to affect autoantibody reactivities through change in the ganglioside epitope on the LOS outer core, resulting in the diverse neurologic features shown by patients with GBS. In this study we used 105 C jejuni isolates from patients with GBS or a clinical variant and compared cst-II gene frequency in the neuropathic and enteritic strains. We also examined the hypothesis that the genetic polymorphism of the bacterium produces the differences in the clinical manifestation of GBS. Methods. Bacterial strains and patients. Since 1990, we have received more than 8,000 requests from Japanese physicians to test serum anti-ganglioside antibodies from patients presenting with various neurologic disorders. On receipt of serum samples from patients with GBS or a clinical variant, we request the primary physicians to do a stool culture and to send the patient’s stool specimen to the Tokyo Metropolitan Institute of Public Health for C jejuni isolation. From 1990 to 2002, 81 C jejuni strains were isolated successfully by one of the authors (M.T.), and 24 strains were obtained from hospitals. These were used in this study (n ⫽ 105: GBS, 80; FS, 20; Bickerstaff brainstem encephalitis,16 3; acute ophthalmoparesis,17 1; acute oropharyngeal palsy,18 1). Most of the strains used were included in our previous study.19 Two, OH4384 and OH4382, were obtained from sibling GBS patients6,20; the others were from sporadic cases with geographically equal distributions. A questionnaire was filled in by the primary physician as to whether diagnostic criteria for GBS or a clinical variant were fulfilled. We also reviewed the patients’ medical records to ascertain diagnoses and neurologic findings. GBS, FS, Bickerstaff brainstem encephalitis, and acute ophthalmoparesis diagnoses were all based on clinical criteria.21,22 ELISAs were used to test for the presence of IgG antibodies to GM1, GD1a, and GQ1b, as reported elsewhere.23 Sixty-five strains that had been isolated from patients with uncomplicated enteritis and collected throughout Japan were the controls. Anti-ganglioside antibodies were not tested in the enteritis patients because previous studies showed that such patients did not have the autoantibodies.24,25 Detection of ganglioside epitopes on LOS. Crude LOS fractions were prepared from the isolates, as described elsewhere,26 with minor modifications.27 We first performed sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis and silver staining to ensure LOS had been extracted then used western blotting with the cholera toxin B-subunit (a ligand for GM1oligosaccharide structure) to examine whether the GM1 epitope was present on it. A 5 ␮L portion of the bacterial lysate was added to each well, after which the samples were separated on 15% tricine-SDS-polyacrylamide gels (SPU-15S series, Atto Corporation, Tokyo, Japan).28 After electrophoresis, the LOS samples were blotted on polyvinylidene difluoride membranes (Atto Corporation), and the membranes incubated at 4 °C for 2 hours with the peroxidase-conjugated cholera toxin B-subunit (List Biologic Laboratories, Campbell, CA) diluted 1:2,000 in phosphate-buffered saline containing 0.5% casein. Binding was made visible with 4-chloro-1-naphtol (Konica Immunostaining HRP-1000, Konica, Tokyo, Japan). Ganglioside-like LOS also was investigated by thin-layer chromatography (TLC) with immunostaining because this method decreases the volume of reagent needed in the immunostaining step. The reagents used are the monoclonal antibodies (GB1 [antiGD1a], GB2 [anti-GM1], and FS1 [anti-GQ1b]),8,27 and sera from patients with GBS (S6960 [anti-GM1] and S5174 [anti-GD1a]) or FS (S7577 [anti-GQ1b]). A 10 ␮L portion of each bacterial lysate was spotted on a precoated Silica Gel 60 TLC plate (Merck, Darmstadt, Germany), developed with an n-propanol-water-25% ammonia solution (6:5:1, by volume), after which the plates were immunostained as reported elsewhere.23 The TLC analysis was done by one of the authors (M.K.). PCR and DNA sequencing. A single bacterial colony was suspended in 300 ␮L of sterile deionized distilled water and boiled for 10 minutes. After centrifugation at 15,000 rpm for 1 minute, the supernatant was used as the template in the PCR amplification. Oligonucleotide primers were designed based on the reported C jejuni cst-II sequence (GenBank accession no. AF400048): forward directional primer, 17F (5=-TTTCTGGAAATGGACCAAGTTT-3=), and reverse, 220R (5=-CGGTCTCATATTCTTGATTTTGG-3=). These primers amplify the 204 bp fragment of the cst-II gene carrying the 51st codon. Amplification reactions were run in a total volume of 50 ␮L with 20 pmol of each primer, 10 ␮L of template, and 2.5 units of Taq DNA polymerase (TaKaRa Ex Taq, Takara Bio Inc., Shiga, Japan), 40 nmol dNTPs, and buffer (2 mM Mg2⫹), as recommended by the manufacturer. After a denaturation step of 5 minutes at 95 °C, the amplification mixture underwent 30 cycles of amplification, each cycle consisting of 10 seconds at 94 °C, 10 seconds at 55 °C, and 30 seconds at 72 °C, in a GeneAmp PCR System 9700 (PE Biosystems, Foster City, CA). PCR products were purified with a QIAquick PCR Purification Kit (Qiagen K.K., Tokyo, Japan). Nucleotide sequences of PCR products were determined with an ABI BigDye Terminator v1.1 Cycle Sequencing kit (Applied Biosystems, Foster City, CA) and an ABI Prism 3100 Genetic Analyzer (Applied Biosystems). Statistical analysis. Frequency differences between the groups were compared by means of Fisher’s exact test using SPSS 12.0J software (SPSS Inc., Chicago, IL). A difference was considered significant when the two-sided p value was less than 0.05. Results. cst-II Gene frequency and polymorphism (Thr/ Asn51). PCR and direct nucleotide sequence analyses showed that the cst-II gene more often was present in neuropathic than enteritic strains (table 1). Although the 51st codon of cst-II is reported to be AAT (corresponding to Asn) or ACC (Thr),15 some strains in our study had AAC (Asn). This indicates that a single base change (A to C or C to A) which may occur during infection can alter the substrate specificity of Cst-II (monofunctional or bifunctional). cst-II (Thr51) was more common in the neuropathic (50%) than enteritic (25%) strains (p ⫽ 0.001), whereas cst-II (Asn51) did not differ (34% vs 26%; p ⫽ 0.31). Also, 9 of 16 November (1 of 2) 2005 NEUROLOGY 65 1377 Table 1 Comparison of genetic and phenotypic properties of neuropathic and enteritic Campylobacter jejuni strains Neuropathic strains, n ⫽ 105 Enteritic strains, n ⫽ 65 p Value cst-II 89 (85) 33 (51) ⬍0.001 cst-III 9 (9) 15 (23) 0.012 Cholera toxin 62 (59) 25 (38) 0.01 GB2 63 (60) 25 (38) 0.007 S6960 60 (57) 22 (34) 0.004 GB1 51 (49) 13 (20) ⬍0.001 S5174 49 (47) 12 (18) ⬍0.001 FS1 22 (21) 13 (20) 1.0 S7577 32 (30) 17 (26) 0.60 Gene GM1 epitope* Figure. The ganglioside-like lipo-oligosaccharide of Campylobacter jejuni and anti-ganglioside autoantibody reactivity in patients with Guillain–Barré syndrome or a variant. Frequencies of positive IgG antibodies against GM1 (left), GD1a (middle), and GQ1b (right) are compared between patients whose isolates carry ganglioside epitope on lipo-oligosaccharide (LOS) (pale bars) or not (dark bars). *p ⫽ 0.04; **p ⬍ 0.001. GD1a epitope* GQ1b epitope* Values are n (%). epitopes coexisted in five strains. Seventeen (16%) of the strains had none of the ganglioside epitopes examined. cst-II Polymorphism (Thr/Asn51) was closely related to the ganglioside epitopes on LOS in both the neuropathic and enteritic strains. Asn51 strains more often expressed GQ1b epitope than did the others (table 2). In contrast, most Thr51 strains carried GM1- and GD1a-like LOS. Because the 53rd codon of cst-II is reported to affect ␣-2,3and ␣-2,8-sialyltransferase activities in an Asn51 strain,15 we analyzed its association with ganglioside-like LOS. Of the seven strains with cst-II (Asn51/Ile53) thought to have relatively low ␣-2,3- and ␣-2,8-sialyltransferase activities,15 four had both GQ1b and GM1 epitopes, three had none. In contrast, all 22 strains (Asn51/Ser53 and Asn51/ Gly53) thought to have high ␣-2,8-sialyltransferase activity had only the GQ1b epitope. Four of seven strains (Asn51/ Tyr53) also had only the GQ1b epitope. The other three had neither epitope. Serologic and neurologic features. Detailed neurologic information was available for 97 neuropathy patients, and pretreatment serum samples were obtained within 4 weeks of neuropathic onset for 94. Serum anti-ganglioside autoantibody reactivities overall corresponded to the ganglioside epitopes on the LOSs of the isolates (figure): GM1 epitope on LOS and anti-GM1 IgG (p value ⬍ 0.001); * The reagents used are the monoclonal antibodies (GB1 [antiGD1a], GB2 [anti-GM1], and FS1 [anti-GQ1b]), and sera from patients with Guillain-Barré syndrome (S6960 [anti-GM1] and S5174 [anti-GD1a]) or Fisher syndrome (S7577 [anti-GQ1b]). Cholera toxin (B-subunit) is a ligand for GM1-oligosaccharide structure. neuropathic strains without cst-II had the cst-III gene,12 and this gene was significantly less frequent in the neuropathic strains compared to the enteritic ones (see table 1). Ganglioside-like LOS. Although we used two or three reagents in the detection of each ganglioside epitope, the overall results were identical, except for some discrepancies probably due to differences in the sensitivities of the reagents (see table 1). Figure E-2 shows the ganglioside epitope detection in representative strains. Neuropathic strains more commonly expressed GM1 and GD1a epitopes than did enteritic strains. In contrast, the frequency of the GQ1b epitope did not differ between them. Immunostaining results for the patients’ sera showed that the GM1 and GD1a epitopes commonly were present in the same neuropathic strains (n ⫽ 48; 46%), whereas the GQ1b epitope was present isolatedly (n ⫽ 27; 26%). GQ1b and GM1 Table 2 Association of bacterial properties with cst-II content and polymorphism in neuropathic Campylobacter jejuni strains cst-II cst-II (Asn51) Present Absent p Value Lipo-oligosaccharide n ⫽ 89 n ⫽ 16 GM1 epitope* 53 (60) 7 (44) 0.28 GD1a epitope* 48 (54) 1 (6) ⬍0.001 GQ1b epitope* 31 (35) 1 (6) 0.04 Present Absent n ⫽ 36 n ⫽ 69 4 (11) 56 (81) 0 49 (71) 30 (83) 2 (3) cst-II (Thr51) p Value Present Absent p Value n ⫽ 53 n ⫽ 52 ⬍0.001 49 (92) 11 (21) ⬍0.001 ⬍0.001 48 (91) 1 (2) ⬍0.001 ⬍0.001 1 (2) 31 (60) ⬍0.001 Values are n (%). * Judged by results of thin-layer chromatography with immunostaining of patients’ sera (S6960 [anti-GM1], S5174 [anti-GD1a], and S7577 [anti-GQ1b]) having high anti-ganglioside antibody titer. 1378 NEUROLOGY 65 November (1 of 2) 2005 Table 3 Association of patients’ clinical features with cst-II content and polymorphism of their Campylobacter jejuni isolates cst-II IgG antibody to Present Absent n ⫽ 80 n ⫽ 14 cst-II (Asn51) p Value Present Absent n ⫽ 32 n ⫽ 62) cst-II (Thr51) p Value Present Absent n ⫽ 48 n ⫽ 46 p Value GM1 49 (61) 9 (64) 1.0 7 (22) 51 (82) ⬍0.001 42 (88) 16 (35) ⬍0.001 GD1a 32 (40) 4 (29) 0.56 7 (22) 29 (47) 0.03 25 (52) 11 (24) 0.006 20 (25) 3 (21) 1.0 18 (56) 5 (8) ⬍0.001 2 (4) 21 (46) ⬍0.001 n ⫽ 81 n ⫽ 16 n ⫽ 33 n ⫽ 64 n ⫽ 48 n ⫽ 49 GQ1b Neurological sign Ophthalmoparesis 25 (31) 4 (25) 0.77 21 (64) 8 (13) ⬍0.001 4 (8) 25 (51) ⬍0.001 Facial palsy 14 (17) 1 (6) 0.45 11 (33) 4 (6) 0.001 3 (6) 12 (24) 0.02 Bulbar palsy 17 (21) 0 0.07 13 (39) 4 (6) ⬍0.001 4 (8) 13 (27) 0.03 Limb weakness 69 (85) 13 (81) 0.71 22 (67) 60 (94) 0.001 47 (98) 35 (71) ⬍0.001 Ataxia 17 (21) 4 (25) 0.74 14 (42) 7 (11) 0.001 3 (6) 18 (37) ⬍0.001 Sensory disturbance 26 (32) 3 (19) 0.38 9 (27) 20 (31) 0.82 17 (35) 12 (24) 0.27 n ⫽ 89 n ⫽ 16 n ⫽ 36 n ⫽ 69 n ⫽ 53 n ⫽ 52) Guillain–Barré syndrome 68 (76) 12 (75) 1.0 17 (47) 63 (91) ⬍0.001 51 (96) 29 (56) ⬍0.001 Fisher syndrome 16 (18) 4 (25) 0.50 14 (39) 6 (9) ⬍0.001 2 (4) 18 (35) ⬍0.001 Diagnosis Values are n (%). GD1a epitope and anti-GD1a IgG (p value ⫽ 0.04); and GQ1b epitope and anti-GQ1b IgG (p ⬍ 0.001). As expected, cst-II polymorphism was closely related to serum anti-ganglioside autoantibody reactivities, and therefore to neurologic features, whereas gene content was not (table 3). Patients from whom C jejuni (Asn51) had been isolated more often had serum anti-GQ1b IgG autoantibody, cranial nerve palsies, and ataxia. In contrast, C jejuni (Thr51) was associated with anti-GM1 and antiGD1a IgG antibodies and limb weakness. The diagnosis was GBS for most patients with C jejuni (Thr51), whereas it was GBS for 47% and FS for 39% of patients with C jejuni (Asn51). Conversely, compared to the enteritis patients, FS patients more often had been infected by C jejuni (Asn51) (70% vs 26%; p value ⫽ 0.001), whereas GBS patients had been by C jejuni (Thr51) (64% vs 25%; p value ⬍ 0.001). Variation in the 53rd codon was not associated with any type of autoantibody or neurologic feature in spite of its association with ganglioside-like LOS (data not shown). Discussion. The pathogenesis of many postinfectious disorders is still unknown, no autoantigens or virulence factors having been identified. For example, why some group A streptococci can cause acute rheumatic fever and others acute glomerulonephritis is unclear.29 In contrast, we are the first to show the detailed molecular mechanism of GBS after C jejuni enteritis, based on our and others findings that the genetic polymorphism of the bacterium alters the substrate specificity of the LOS biosynthesis enzyme15 and that autoantibody reactivity determines the clinical presentation of GBS.30 These findings suggest that the genetic polymorphism of antecedent agents determines autoantibody reactivities and clinical manifestations through change to the host-mimicking molecule in some post-infectious disorders. We believe that GBS is the first paradigm to explain the detailed pathogenesis of a postinfectious, autoimmune-mediated, molecular mimicrytriggering disorder. We confirmed results of a previous report that cst-II more frequently exists in GBS than in enteritis isolates, indicative that its presence is a risk factor for developing GBS.14 However, considerable numbers of strains from patients with uncomplicated enteritis also had cst-II gene, indicating that cst-II gene is necessary but not adequate for initiating autoimmune response, although cst-II genotype is important in determining antibody reactivity when autoimmune response is triggered. Certain other genes (e.g., cst-III12 or an unidentified gene) might produce the enzyme protein instead, subsequently sialylating LOS. Three genes, cst-I, -II, and –III, are reported to encode sialyltransferase protein,12,15 but cst-I was lacking in some strains with sialylated LOS and therefore is unlikely to be responsible for LOS sialylation.10 Because Cst-III appears to have only ␣-2,3-sialyltransferase activity (monofunction),15 cst-II content must be essential for ␣-2,8sialyltransferase activity and thereby biosynthesis of the GQ1b epitope.13,15 Most of the previous studies have failed to find a specific C jejuni genotype for GBS and FS.31-35 It was recently reported that the class A LOS biosynthesis locus was over-represented in GBS-associated as compared to enteritis strains, whereas all four of the FS-related strains belonged to class B.36 The authors suspected that the frequent expression of a GM1 epitope in class A and a GQ1b epitope in class B strains is responsible for the development of GBS and FS. Their findings, however, do not provide the answers as to which difference leads to diverse November (1 of 2) 2005 NEUROLOGY 65 1379 ganglioside-mimics (GM1 and GQ1b) in spite of there being almost the same class A and B gene profiles.15 In contrast, our data clearly indicate that both presence and polymorphism of bacterial cst-II have a major role in the type of ganglioside-like structure on LOS, thereby determining autoantibody reactivity and the pattern of neurologic presentation. Phase variation owing to a homopolymeric G-tract in cst-II or other LOS synthesis genes also may be related to what ganglioside epitopes are present.10 Along with those of a previous study,15 however, our findings suggest that mainly it is variation in the cst-II 51st codon and supplemental variation in the 53rd codon that determine which ganglioside epitopes are present on LOS, but only the former was related to the autoantibody reactivity in and neurologic features of neuropathy patients. Interestingly, Haemophilus influenzae, a pathogen recently suggested to cause GBS and FS,27,37 also has sialylated LOS.38 Three genes (lic3A, siaA, lsgB) have been cloned for that sialylation enzyme.39,40 Whether the polymorphism of H influenzae genes also is related to autoantibody reactivity and consequently to the neurologic features of GBS and FS requires investigation. C jejuni strains which had been isolated from anti-GQ1b antibody-positive patients often carried GQ1b epitope on the LOS.41 The specificity of antiganglioside antibody induced by immunization of C jejuni LOS overall corresponds to ganglioside epitopes on the LOS in rabbits.42 In this study, we showed that the target ganglioside (GM1, GD1a, or GQ1b) for serum autoantibody corresponds to the LOS-mimicking gangliosides of isolates from individual patients. However, other gangliosides such as GM1b and GalNAc-GD1a could be target antigens for autoantibodies in GBS30 and further investigation is necessary to explain the variety of the clinical manifestation of GBS in more detail. Acknowledgment The authors thank Dr. Michel Gilbert (Institute for Biologic Sciences, National Research Council of Canada, Ottawa, Canada) for critical reading of the paper; Saiko Koike and Takashi Namatame (Institute for Medical Science, Dokkyo University School of Medicine, Tochigi, Japan) for help with the bacterial and sequence analyses; Maki Okazaki (Department of Neurology, Dokkyo University School of Medicine, Tochigi, Japan) for technical assistance; and Dr. Yukihiro Nishimoto (Department of Pediatrics, Kinan General Hospital, Wakayama, Japan) and Dr. Shigeru Matsushita (Tama Branch Laboratory, Tokyo Metropolitan Institute of Public Health, Tokyo, Japan) for providing the C jejuni strains from enteritis patients. References 1. Ropper AH. The Guillain-Barré syndrome. N Engl J Med 1992;326: 1130–1136. 2. Ogawara K, Kuwabara S, Mori M, et al. Axonal Guillain-Barré syndrome: relation to anti-ganglioside antibodies and Campylobacter jejuni infection in Japan. Ann Neurol 2000;48:624–631. 3. Chiba A, Kusunoki S, Obata H, et al. Serum anti-GQ1b IgG antibody is associated with ophthalmoplegia in Miller Fisher syndrome and Guillain-Barré syndrome: clinical and immunohistochemical studies. Neurology 1993;43:1911–1917. 4. Jacobs BC, Rothbarth PH, van der Meché FGA, et al. The spectrum of antecedent infections in Guillain-Barré syndrome: a case-control study. Neurology 1998;51:1110–1115. 1380 NEUROLOGY 65 November (1 of 2) 2005 5. Yuki N, Taki T, Inagaki F, et al. A bacterium lipopolysaccharide that elicits Guillain-Barré syndrome has a GM1 ganglioside-like structure. J Exp Med 1993;178:1771–1775. 6. Aspinall GO, McDonald AG, Pang H, et al. Lipopolysaccharides of Campylobacter jejuni serotype O:19 structures of core oligosaccharide regions from the serostrain and two bacterial isolates from patients with Guillain-Barré syndrome. Biochemistry 1994;33:241–249. 7. Shin JE, Sckloo S, Mainkar, et al. Lipo-oligosaccharides of Campylobacter jejuni serotype O:10: structures of core oligosaccharide regions from a bacterial isolate from a patient with the Miller-Fisher [sic] syndrome and from the serotype reference strain. Carbohydr Res 1998; 305:223–232. 8. Yuki N, Susuki K, Koga M, et al. Carbohydrate mimicry between human ganglioside GM1 and Campylobacter jejuni lipo-oligosaccharide causes Guillain-Barré syndrome. Proc Natl Acad Sci USA 2004;101: 11404–11409. 9. Parkhill J, Wren BW, Mungall K, et al. The genome sequence of the food-borne pathogen Campylobacter jejuni reveals hypervariable sequences. Nature 2000;403:665–668. 10. Gilbert M, Brisson J-R, Karwaski M-F, et al. Biosynthesis of ganglioside mimics in Campylobacter jejuni OH4384: identification of the glycosyltransferase genes, enzymatic synthesis of model compounds, and characterization of nanomole amounts by 600-MHz 1H and 13C NMR analysis. J Biol Chem 2000; 275: 3896–3906. 11. Svennerholm L. Chromatographic separation of human brain gangliosides. J Neurochem 1963;10:613–623. 12. Guerry P, Ewing CP, Hickey TE, et al. Sialylation of lipooligosaccharide cores affects immunogenicity and serum resistance of Campylobacter jejuni. Infect Immun 2000;68:6656–6662. 13. van Belkum A, van den Braak N, Godschalk P. Campylobacter jejuni gene associated with immune-mediated neuropathy. Nat Med 2001;7: 752–753. 14. Nachamkin I, Liu J, Li M, et al. Campylobacter jejuni from patients with Guillain-Barré syndrome preferentially expresses a GD1a-like epitope. Infect Immun 2002;70:5299–5303. 15. Gilbert M, Karwaski M-F, Bernatchez S, et al. The genetic bases for the variation in the lipo-oligosaccharide of the mucosal pathogen, Campylobacter jejuni: biosynthesis of sialylated ganglioside mimics in the core oligosaccharide. J Biol Chem 2002;277:327–337. 16. Odaka M, Yuki N, Yamada M, et al. Bickerstaff’s brainstem encephalitis: clinical features of 62 cases and a subgroup associated with Guillain-Barré syndrome. Brain 2003;126:2279–2290. 17. Yuki N, Odaka M, Hirata K. Acute ophthalmoparesis (without ataxia) associated with anti-GQ1b IgG antibody: clinical features. Ophthalmology 2001;108:196–200. 18. O’Leary CP, Veitch J, Durward WF, et al. Acute oropharyngeal palsy is associated with antibodies to GQ1b and GT1a gangliosides. J Neurol Neurosurg Psychiatry 1996;61:649–651. 19. Takahashi M, Koga M, Yokoyama K, Yuki N. Epidemiology of Campylobacter jejuni isolated from patients with Guillain-Barré and Fisher syndromes in Japan. J Clin Microbiol 2005;43:335–339. 20. Yuki N, Tsujino Y. Familial Guillain-Barré syndrome subsequent to Campylobacter jejuni enteritis. J Pediatr 1995;126:162. 21. Asbury AK, Cornblath DR. Assessment of current diagnostic criteria for Guillain-Barré syndrome. Ann Neurol 1990;27(suppl):S21–24. 22. Odaka M, Yuki N, Hirata K. Anti-GQ1b IgG antibody syndrome: clinical and immunological range. J Neurol Neurosurg Psychiatry 2001;70: 50–55. 23. Yuki N, Tagawa Y, Irie F, et al. Close association of Guillain-Barré syndrome with antibodies to minor monosialogangliosides GM1b and GM1␣. J Neuroimmunol 1997;74:30–34. 24. Yuki N, Yoshino H, Sato S, Miyatake T. Acute axonal polyneuropathy associated with anti-GM1 antibodies following Campylobacter enteritis. Neurology 1990;40:1900–1902. 25. Rees JH, Gregson NA, Hughes RAC. Anti-ganglioside GM1 antibodies in Guillain-Barré syndrome and their relationship to Campylobacter jejuni infection. Ann Neurol 1995;38:809–816. 26. Hitchcock PJ, Brown TM. Morphological heterogeneity among Salmonella lipopolysaccharide chemotypes in silver-stained polyacrylamide gels. J Bacteriol 1983;154:269–277. 27. Koga M, Gilbert M, Li J, et al. Antecedent infections in Fisher syndrome. A common pathogenesis of molecular mimicry. Neurology 2005; 64:1605–1611. 28. Schägger H, von Jagow G. Tricine-sodium dodecyl sulfatepolyacrylamide gel electrophoresis for the separation of proteins in the range from 1 to100 kDa. Anal Biochem 1987;166:368–379. 29. Stollerman GH. Rheumatic fever. Lancet 1997;349:935–942. 30. Willison HJ, Yuki N. Peripheral neuropathies and anti-glycolipid antibodies. Brain 2002;125:2591–2625. 31. Endtz HP, Ang CW, van den Braak N, et al. Molecular characterization of Campylobacter jejuni from patients with Guillain-Barré and Miller Fisher syndromes. J Clin Microbiol 2000;38:2297–2301. 32. Duim B, Ang CW, van Belkum A, et al. Amplified fragment length polymorphism analysis of Campylobacter jejuni strains isolated from chickens and from patients with gastroenteritis or Guillain-Barré or Miller Fisher syndromes. Appl Environ Microbiol 2000;66:3917–3923. 33. Engberg J, Nachamkin I, Fussing V, et al. Absence of clonality of Campylobacter jejuni in serotypes other than HS:19 associated with Guillain-Barré syndrome and gastroenteritis. J Infect Dis 2001;184: 215–220. 34. Nachamkin I, Engberg J, Gutacker M, et al. Molecular population genetic analysis of Campylobacter jejuni HS:19 associated with GuillainBarré syndrome and gastroenteritis. J Infect Dis 2001;184:221–226. 35. Leonard II EE, Tompkins LS, Falkow S, Nachamkin I. Comparison of Campylobacter jejuni isolates implicated in Guillain-Barré syndrome and strains that cause enteritis by a DNA microarray. Infect Immun 2004;72:1199–1203. 36. Godschalk PCR, Heikema AP, Gilbert M, et al. The crucial role of Campylobacter jejuni genes in autoimmune antibody induction. J Clin Invest 2004;114:1659–1665. 37. Mori M, Kuwabara S, Miyake M, et al. Haemophilus influenzae infection and Guillain-Barré syndrome. Brain 2000;123:2171–2178. 38. Mandrell RE, McLaughlin R, Abu Kwaik Y, et al. Lipooligosaccharides (LOS) of some Haemophilus species mimic human glycosphingolipids, and some LOS are sialylated. Infect Immun 1992;60:1322–1328. 39. Hood DW, Cox AD, Gilbert M, et al. Identification of a lipopolysaccharide ␣-2,3-sialyltransferase from Haemophilus influenzae. Mol Microbiol 2001;39:341–350. 40. Jones PA, Samuels NM, Phillips NJ, et al. Haemophilus influenzae type b strain A2 has multiple sialyltransferases involved in lipooligosaccharide sialylation. J Biol Chem 2002;277:14598–14611. 41. Ang CW, Laman JD, Willison HJ, et al. Structure of Campylobacter jejuni lipopolysaccharides determines antiganglioside specificity and clinical features of Guillain-Barré and Miller Fisher patients. Infect Immun 2002;70:1202–1208. 42. Ang CW, Noordzij PG, de Klerk MA, et al. Ganglioside mimicry of Campylobacter jejuni lipopolysaccharides determines antiganglioside specificity in rabbits. Infect Immun 2002;70:5081–5085. NeuroImages Isolated insular infarction eliminates contralateral cold, cold pain, and pinprick perception Frank Birklein, MD, PhD; Roman Rolke, MD; and Wibke Müller-Forell, MD, PhD, Mainz, Germany Functional imaging suggests that the insular cortex is important for pain processing.1 We performed quantitative sensory testing (QST) in a 64-year-old man with acute left insular infarct. At day 5, diffusion-weighted MRI (figure) and QST (warm, cold, heat, and cold pain; tactile, vibratory, pinprick, and pressure pain) were performed. Contralateral to the infarct, the patient did not feel cold (⌬32 °C), cold pain (⌬32 °C), or pinprick pain (⬎630 mN). The other sensory modalities were normal. There were no other neurologic disturbances apart from a mild receptive aphasia. This case illustrates that isolated insular infarction indeed leads to sensory disturbances,2 which might even be modality specific. Copyright © 2005 by AAN Enterprises, Inc. 1. Craig AD, Chen K, Bandy D, Reiman EM. Thermosensory activation of insular cortex. Nat Neurosci 2000;3:184–190. 2. Bassetti C, Bogousslavsky J, Regli F. Sensory syndromes in parietal stroke. Neurology 1993;43:1942–1949. Figure. Diffusion-weighted MR image shows the acute ischemic infarct, which is restricted to the middle/posterior left insular cortex. Address correspondence and reprint requests to Dr. F. Birklein, Department of Neurology, University of Mainz, Langenbeckstrasse 1, D-55101 Mainz, Germany; e-mail: birklein@neurologie.klinik.uni-mainz.de November (1 of 2) 2005 NEUROLOGY 65 1381 Isolated insular infarction eliminates contralateral cold, cold pain, and pinprick perception Frank Birklein, Roman Rolke and Wibke Müller-Forell Neurology 2005;65;1381 DOI 10.1212/01.wnl.0000181351.82772.b3 This information is current as of November 7, 2005 Updated Information & Services including high resolution figures, can be found at: http://www.neurology.org/content/65/9/1381.full.html References This article cites 2 articles, 1 of which you can access for free at: http://www.neurology.org/content/65/9/1381.full.html##ref-list-1 Citations This article has been cited by 4 HighWire-hosted articles: http://www.neurology.org/content/65/9/1381.full.html##otherarticles Permissions & Licensing Information about reproducing this article in parts (figures,tables) or in its entirety can be found online at: http://www.neurology.org/misc/about.xhtml#permissions Reprints Information about ordering reprints can be found online: http://www.neurology.org/misc/addir.xhtml#reprintsus Neurology ® is the official journal of the American Academy of Neurology. Published continuously since 1951, it is now a weekly with 48 issues per year. Copyright . All rights reserved. Print ISSN: 0028-3878. Online ISSN: 1526-632X.