You are on page 1of 7

Journal of the Neurological Sciences 196 (2002) 1 – 7

www.elsevier.com/locate/jns

Review article
Sialosyl-galactose: a common denominator of Guillain–Barré
and related disorders?
Anthony P. Moran a,*, Martina M. Prendergast a, Edward L. Hogan a,b
a
Department of Microbiology, National University of Ireland, Galway, Ireland
b
Department of Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
Received 6 September 2001; received in revised form 20 December 2001; accepted 5 February 2002

Abstract

The immune reactivity implicated in the pathogenesis of Guillain – Barré syndrome (GBS) and related diseases, which occur following
infection with specific strains of Campylobacter jejuni bearing sialylated lipopolysaccharide structures that cross-react with specific
gangliosides, is consistent with provocation of inflammation via molecular mimicry. In this review, we have focused upon microbial
characteristics and structures, the fine structure of the essential carbohydrate determinants, and the application of our proposed criteria,
modified from those of Koch for causation of infectious and of Witebsky for autoimmune diseases, to the circumstance of infectious
induction of autoimmune disorder. D 2002 Elsevier Science B.V. All rights reserved.

Keywords: Guillain – Barré syndrome; Miller Fisher syndrome; Campylobacter jejuni; Lipopolysaccharides; Anti-ganglioside antibodies; Autoimmune disease;
Peripheral nervous system disorders

1. Perspective: Peripheral nervous system (PNS) with plasmapheresis or intravenous high dose immunoglo-
disorder, clinical phenotype, and anti-ganglioside bulin therapy [6– 8].
antibodies Autoimmune disorders of PNS similar to GBS include a
neuropathy induced by parenteral GM1 therapy [9], and
The recent advances in insight into the pathogenesis of multifocal motor neuropathy (MMN) in which high propor-
Guillain – Barré syndrome (GBS) includes recognition of the tions of anti-GM1 antibodies occur in many patients (range,
frequency of preceding infection with Campylobacter 22 – 85%) [10]. The wide range of anti-ganglioside antibody
jejuni, the frequency of elevated serum titers to putatively specificities reported in GBS contrasts with the limited range
neuritogenic gangliosides, and the findings of elevated and of antibody specificities found in MMN [11], and the
specific serum titers to the lipopolysaccharide or lipooligo- disorder caused by parenteral ganglioside therapy. In our
saccharide (LPS/LOS) of certain strains of C. jejuni [1– 4]. literature survey [1], the 44 reports included multiple find-
The spectrum of clinical syndromes ranges from classical, ings of anti-ganglioside antibodies to GM1, GM1b, GQ1b,
largely motor, acute inflammatory demyelinating polyneur- LM1, GalNAc-GD1a, and less frequent findings of antibodies
opathy (AIDP) to a purely motor form called acute motor to GM2, GD1a, GD1b, GT1b, and GA1 (asialo-GM1). Anti-
axonal neuropathy (AMAN), acute sensory neuropathy GQ1b antibodies are found in a high percentage of patients
(ASN), and acute motor and sensory axonal neuropathy with MFS (up to 100% of cases) [2]. In contrast to the IgM
(AMSAN) [3] and to the Miller Fisher syndrome (MFS) isotype of anti-GM1 antibody found in chronic neuropathies,
manifesting ophthalmoplegia, ataxia, and areflexia [5]. Suc- GBS sera contains IgG or less commonly IgA isotypes [12].
cessful therapy for these disorders has often been obtained It is noteworthy that the elevated antibody titers in these
chronic neuropathies often decline and disappear during
recovery.
The correlation of anti-ganglioside antibody specificities
*
Corresponding author. Tel.: +353-91-524411x3163; fax: +353-91-
with clinical subtypes of GBS are strongest for the associ-
525700. ations of anti-GD1a and anti-GalNAc-GD1a with AMAN
E-mail address: anthonymoran@nuigalway.ie (A.P. Moran). [4,13] although one report disagreed [14], of anti-GM1b

0022-510X/02/$ - see front matter D 2002 Elsevier Science B.V. All rights reserved.
PII: S 0 0 2 2 - 5 1 0 X ( 0 2 ) 0 0 0 3 6 - 9
2 A.P. Moran et al. / Journal of the Neurological Sciences 196 (2002) 1–7

with AMAN in one Japanese study [15], of anti-GQ1b with in nature, but rather capsular polysaccharide (CPS), a
MFS [2], and of anti-GT1a with oropharyngeal palsy [16]. In molecule not previously described in C. jejuni, arose after
most instances, there is cross-reactivity of serum antibodies the identification of a cluster of genes with significant
with several gangliosides. sequence similarity to the capsular PS genes (kps genes)
of E. coli [31,32]. Interestingly, we have previously chemi-
cally characterized LPS/LOS-independent PS from C. jejuni
2. Antecedent C. jejuni infections, LPS/LOS, and GBS HS:3 and HS:41 [34,35] although the role of the PSs in
serotyping was not determined. Determining the role of
C. jejuni is the most common infectious precursor of GBS LPS/LOS in serotyping has attracted renewed interest
occurring in 32% of GBS patients overall, whereas other because serotyping of clinical isolates has confirmed an
antecedent pathogens include cytomegalovirus (CMV) association between certain serotypes of C. jejuni and
[17,18], Epstein –Barr virus (EBV) [19], and mycoplasma subsequent development of GBS [1]. In general, LPS has
[19,20]. Hepatitis A and B, varicella zoster virus, human three components: the hydrophobic, membrane-inserted
immunodeficiency virus, and herpes simplex virus are linked lipid A, an oligosaccharide (OS) with inner and outer
by more equivocal data with similar prevalences in the components, and the externally directed polysaccharide O
controls [21]. C. jejuni infection is associated with more chain, whereas LOS lacks the O-chain moiety [12]. C. jejuni
severe GBS and greater residual disability [22]. The fre- LPS/LOS is more complex than that of comparable enteric
quency of C. jejuni infection in GBS ranges from 14– 66% pathogens such as Salmonella in that the core region is
(see Ref. [1]), with an overall prevalence of 32% as shown in structurally diverse, and sufficiently so to contribute to
Table 1. The duration of excretion of C. jejuni is brief (mean differences in serospecificity [36]. Importantly, sialic acid
of 16 days) [23], and the majority of patient stools have is present in the core OS of certain C. jejuni strains [37],
cleared the bacterium by the onset of symptomatic GBS 1 – 3 particularly those of strains that resemble gangliosides
weeks after the onset of diarrhea [24]. Serological assessment [1,36,46]. In resolving the molecular basis of the heat-stable
is a more sensitive marker for detecting the presence of C. antigen serotyping scheme, we found that LPS/LOS con-
jejuni, but is less specific than stool culture (Table 1). tributes to serospecificity determinant of some strains of C.
C. jejuni is a microaerophilic, gram-negative, non-spore- jejuni, whereas in others, extracellular PS alone is respon-
forming bacterium with a characteristic S-shaped or spiral sible for the heat-stable serotype [47]. Moreover, in other C.
morphology [25]. Infection is zoonotic, and the main route jejuni serotypes, LPS/LOS together with extracellular PS,
of human infection is by ingestion of undercooked poultry contribute to serospecificity. Thus, while both LPS/LOS and
or untreated water and milk [26]. The usual clinical mani- extracellular PS are involved in serotyping, whether only
festation is acute inflammatory enterocolitis [23,27], and one molecule or both together are responsible for serospe-
diarrhea caused by C. jejuni infection exceeds the combined cificity is dependent on the particular C. jejuni serotype.
total for that attributed to Salmonella, Shigella, and Escher- Numerous C. jejuni serotypes have been reported in
ichia coli O157:H7 [28]. C. jejuni infection is unquestion- association with GBS. A strong association was reported
ably underreported: the published incidence rates vary from by Kuroki et al. [38] with a predominance of serotype HS:19,
6/100,000 to 290/100,000. an uncommon serotype in patients with gastroenteritis, in
Serotyping of C. jejuni is based upon differences in the 81% of their GBS population. In addition, Fujimoto et al.
saccharide structure of the bacterial heat-stable antigens, and [39] described four C. jejuni HS:19 isolates from GBS.
strains have been designated as either ‘O’, Penner or heat- Serotype HS:19 was also isolated from GBS patients in
stable (HS) serotypes [29]. Serotyping by passive hemag- Ireland [40], and in the US where it comprised 33% of GBS
glutination (PHA) is determined by exposing typing antisera isolates [41]. Another serotype, C. jejuni HS:41, was isolated
to heated extracts of C. jejuni bound to mammalian red from 6/9 GBS patients in South Africa [42], which is in
blood cells [30]. In the past, the typing scheme has been striking contrast to its overall rarity (only 12/7119 isolates of
considered to be solely based upon LPS/LOS-like mole- C. jejuni). Other C. jejuni serotypes identified in association
cules. However, recent evidence has implicated a role for with GBS include HS:1, HS:2, HS:2/44, HS:4, HS:4/59,
extracellular polysaccharides (PSs) as heat-stable antigens HS:5, HS:10, HS:13, HS:15, HS:16, HS:18, HS:21, HS:24,
as well. Claims that the heat-stable antigen is not LPS/LOS HS:30, HS:37, HS:44, and HS:64 [1]. For MFS, the sero-
types HS:2, HS:10, and HS:23 have been reported [43 – 45].
Table 1 N-acetylneuraminic acid (Neu5Ac) was discovered in C.
Association of C. jejuni infection and GBS (reports)a jejuni strains by Moran et al. [37] in 1991 and subsequent
C. jejuni infection #Infected #GBS Frequency (%) #Reports chemical studies have revealed that structures in the termi-
Stool culture only 108 37 34.3 4
nal regions of the core OS of specific serotypes mimic the
Serology only 1481 489 33.0 18 structures of gangliosides purified from brain of vertebrates
Both stool and culture 113 28 24.8 3 including humans (Table 2). The structural similarity
Total 1702 554 32.6 25 spawned the idea that the immune-mediated response to
a
Summarized from Ref. [1]. C. jejuni epitopes elicits cross-reactive anti-ganglioside
A.P. Moran et al. / Journal of the Neurological Sciences 196 (2002) 1–7 3

Table 2 from a patient we studied [52] who developed GBS after


C. jejuni LPS/ganglioside structural mimicrya
parenteral administration of gangliosides cross-reacted with
Heat-stable serotype Gangliosideb Reference the serotype HS:2, HS:4, HS:19, and HS:41 LPSs and this
HS:1 GM1 [36] was in the absence of a preceding infection with C. jejuni.
HS:2 Several [36] This clearly demonstrates that gangliosides and LPS from C.
HS:4 GD1a [36]
jejuni GBS-associated serotypes are cross-reactive [52].
HS:10 MFS GD3 [45]
HS:19 GD1a, GM1 [46] Moreover, IgM anti-GM1 monoclonal antibodies (MAbs)
HS:19 GBS GT1a [46] from patients with MMN react with LPSs from serotype
HS:19 GBS GD3 [46] HS:4, HS:19, and HS:50 strains [53]. We found GM1-
HS:19 GBS GM1 [40] mimetic epitopes in LPS/LOS of serotype HS:41 GBS
HS:19 GBS GD1a [40]
isolates using a panel of anti-GM1 MAbs cloned from
HS:19 GBS GM1 [40]
HS:23 GM2 [36] patients with MMN [54]. The pattern of binding indicated
HS:23 MFS GD2, GD3, GM2 [47] that there was substantial heterogeneity among the serotype
HS:36 GM2 [36] HS:41 strains in GM1 epitope expression. In this study, it was
HS:41 GBS GM1 [48] first shown that the MAbs reacted exclusively with the core
a
Summarized from Ref. [1]. OS of LPS/LOS and not with lipid A or the polysaccharide
b
Ganglioside that C. jejuni LPS mimics. moiety. In GBS, IgG subclasses are normally restricted to
IgG1 and IgG3, which has been taken to indicate T cell-
antibodies that target neural cells and tissues bearing ganglio- dependent responses to protein antigen. However, we and
sides. However, due to the fact that gangliosides are widely others have observed the production of these IgG subclasses
distributed throughout the nervous system, there should be a in response to glycolipid antigens, including LPS/LOS
high level of B cell tolerance to structurally similar C. jejuni [55,56].
core OS structures. Therefore, tolerance to gangliosides is Furthermore, MFS is of great interest because of the
broken, but the underlying mechanisms of why this happens specific correlation with anti-GQ1b antibodies. There have
have not been addressed experimentally. The supportive been reports of cross-reactivity of GQ1b ganglioside and
findings for the hypothesis of molecular mimicry include LPS/LOSs from C. jejuni HS:2 and HS:23 strains [33,
that the most frequently isolated LPS/LOSs of C. jejuni- 43,44], though it is important to remember that none of
associated GBS are GM1 and GD1a-bearing structures the purified LPS/LOSs from neuropathy-associated strains
[36,40,46,49,50] and also that anti-GM1 antibodies are the have yielded a complete GQ1b structure. The finding of
most frequently observed antibodies in GBS [1]. Other C. GD2/GD3 ganglioside mimicry in a C. jejuni HS:23 strain
jejuni LPS/LOSs bear structural resemblances with GD3, may be relevant because GD3 bears the terminal trisacchar-
GD2, GM2, GM3, and GT1a gangliosides [45,46,48]. The ide Neu5Aca2 ! 8NeuAca2 ! 3Gal, and this is identical
minimal primary structure or ‘‘lowest common denomina- to the terminal of GQ1b ganglioside, a moiety that is
tor’’ for all of these gangliosides is the structure: Neu5- repeated internally [48].
Aca2 ! 3Galh1 ! 4-. How or whether the heterogeneity in The repeated finding of ganglioside epitopes in C. jejuni
ganglioside structure, stemming from variation in sialylation LPS/LOS by cross-reactivity with GBS anti-ganglioside
and in the location on the oligosaccharide of the minimum serum is interpreted then as further justifying the conclusion
sialosyl-galactose unit, i.e., bound proximally or distally, that molecular mimicry determines the neuritogenicity of C.
affects the neuritogenic potency is unclear. However, judg- jejuni.
ing from the cluster epidemiology in northern China and
Japan, and the more severe disability in these GBS patients
[22,50], the relatively undecorated determinant (i.e., GM1
ganglioside) and disialylated determinant (i.e., GD1a or
GalNAc-GD1a ganglioside structures) is the most neurito-
genic. This also applies to correlation of C. jejuni LPS/LOS
structure with clinical GBS severity and derives a corollary
hypothesis that the determinant’s potency increases in the
unmasked, sialosyla2 – 3galactose, and/or doubled, sialo-
syla2 –8sialosyla3 – 2galactose, structures (Fig. 1).
Serological studies are consistent with the mimicry
hypothesis. For example, Oomes et al. [62] demonstrated
that anti-ganglioside antibodies in GBS sera recognized
surface epitopes on whole C. jejuni bacteria in a strain-
specific fashion. With Schwerer et al. [51], we showed that
Fig. 1. Ganglioside core motif associated with Guillain – Barré and Miller
IgA antibodies from C. jejuni-infected patients recognized Fisher syndromes. Schematic representation of gangliosides: Q, galactose;
GM1 and C. jejuni HS:2, HS:4, and HS:19 LPS. Serum IgG E, N-acetylneuraminic acid.
4 A.P. Moran et al. / Journal of the Neurological Sciences 196 (2002) 1–7

3. Postulation of anti-ganglioside pathogenicity in parenteral administration of gangliosides with the C. jejuni


Guillain –Barré syndrome LPS/LOS provide strong corollary evidence [52].
(3) Experimental disease reproduction in an animal host
The proposal that an autoimmune disease is induced by by administration of the tissue target antigen, a molecular
infection requires rigorous criteria for proof. An important mimic, or anti-target antibodies. In this regard, a central
clinical criterion for establishing the existence of an anti- assumption is that there is interspecies conservation of the
body-mediated autoimmune disorder in man is successful tissue target antigen. Antibodies to gangliosides [9,64 – 66]
treatment with reduction of disability attributed to and/or and those raised to C. jejuni LPS/LOS [56] bind to the node
termination of progression of acute neuropathy by removal of Ranvier and to the neuromuscular junction [55,65,67,68].
of circulating antibodies via plasmapheresis, or by intravenous Using experimental in vitro systems to prove that anti-
administration of polyvalent immunoglobulins, where the ganglioside antibodies are relevant factors in GBS patho-
mechanism of action is still unclear [57,58]. Successful genesis has been fraught with difficulties. Exposure of
therapy by both modalities has been found in several clinical isolated nerve preparations to human and rabbit anti-GM1
studies carried out with rigorous criteria for unbiased con- antibodies gives variable results, with some studies reporting
clusions [6 –8,57,58]. acute conduction block of myelinated nerve fibres [69,70],
Criteria for determination of causation are of course whereas other studies report the absence of significant nerve
essential for assessing the validity of proposed biological changes [71]. However, a similar study by this group
mechanisms. Robert Koch’s principles for establishing a role demonstrated that anti-GM1-containing sera from patients
for a pathogen in disease causation requires that: (1) the with MMN blocked conduction in mouse nerves [72]. Most
pathogen be present, (2) be isolated from the host, (3) the reports of intraneural injection of GBS sera into animal
disease be reproduced in an experimental host, and (4) the nerves observe a rapid conduction block and demyelination
pathogen be recovered from the experimental host. They [69,70,73– 77], an effect that appears to be complement-
were extended to autoimmunity by Witebsky et al. [59] in dependent [64,67]. In fact, the presence of complement has
1957 in a study of both Hasimoto’s and experimental auto- been demonstrated at axonal sites and on myelin sheath
immune thyroiditis. They introduced criteria for autoimmun- [78,79], and the presence of IgG antibodies has been shown
ity although not directed at the role of infection [60] that at presynaptic terminals in GBS patients [9,80] and on the
include: (1) demonstration of free circulating antibodies, (2) axon [81]. With regard to MFS, sera with GQ1b antibodies
recognition of the target antigen, (3) production of antibodies induce complement-mediated electrophysiological and im-
in experimental animals against this target antigen, and (4) munological abnormalities [55,64,67]. In contrast, Buch-
the appearance of pathological changes in the corresponding wald et al. [80] demonstrated that these effects could be
tissues of an actively sensitized animal that are basically achieved with anti-GQ1b-negative sera, but state that an IgG
similar to the human disease. antibody factor, not directed against known gangliosides, is
In a similar fashion, we propose a set of rules for responsible for the nerve damage [80,82]. These results
establishing that an autoimmune disorder stems from a suggest that nerve damage in GBS/MFS may not be exclu-
specific infection. These postulates are considered in rela- sively attributed to one determinant. Moreover, the role of
tion to the GBS paradigm in the following. antibody in GBS pathogenesis may be in an antibody-
(1) Antecedent infection is established in the host. We dependent cellular cytotoxicity capacity. Despite these seem-
add the caveats that: (a) the temporal association may vary ingly contradictory results, cumulative evidence supports a
in different infection/autoimmune paradigms, and (b) the role for anti-ganglioside antibodies and complement in GBS
infection may be occult. This is fulfilled in GBS by the pathogenesis, and that the neuromuscular junction and nodes
isolation of C. jejuni, and the high incidence of occurrence of Ranvier are important target sites for the action of humoral
of cross-reactivity of anti-ganglioside antibodies with spe- factors. How T cells contribute to the pathophysiology of
cific C. jejuni LPS/LOSs that are structurally homologous to GBS, and the mechanisms by which they do so is unclear.
gangliosides. Preliminary evidence suggests that C. jejuni LPS/LOS acti-
(2) Cross-reactivity of the pathogen with a host target vates T cell clones by a CD1-dependent mechanism and
molecule should be demonstrated. The serological cross- independent of MHC restriction [83]. Activated T cells may
reactivity of anti-ganglioside antibodies and the ganglioside- produce a nonspecific, cytokine-mediated response, which
mimicking C. jejuni LPS/LOSs show this directly. However, increases the permeability of the blood – nerve barrier allow-
ganglioside-like structures have also been demonstrated in ing access of anti-ganglioside antibodies. Although clinical
C. jejuni strains from patients with uncomplicated enteritis neuropathy has not been produced in experimental animals
[3,40,49,61]. Thus, the humoral response to cross-reactive by passive or active immunization employing either GM1 or
epitopes in C. jejuni LPS/LOS is different in GBS, as C. jejuni LPS/LOSs, it is intriguing that parenteral injection
patients with enteritis rarely develop anti-ganglioside anti- of GD1b causes ‘‘sensory ataxic neuropathy’’ in rabbits
bodies [63]. The cross-reactivity of specific anti-ganglioside [13], and passive transfer of GD1b into rabbits produces
antibodies obtained from patients with MMN [54], and from axonal degeneration and macrophage infiltration resembling
those patients developing polyradiculoneuropathy after AMAN [84]. Recently, there has been a report of the in vivo
A.P. Moran et al. / Journal of the Neurological Sciences 196 (2002) 1–7 5

induction of demyelinating neuropathy after a median inter- [3] Nachamkin I, Allos BM, Ho T. Campylobacter species and Guillain –
Barré syndrome. Clin Microbiol Rev 1998;11:555 – 67.
val of 43 days in rabbits with either a mixture of bovine brain
[4] Ho TW, Willison HJ, Nachamkin I, Li CY, Veitch J, Ung H, et al.
ganglioside or GM1 and both keyhole limpet hemocyanin Anti-GD1a antibody is associated with axonal but not demyelinating
and complete Freund’s adjuvant [81]. This study would, in forms of Guillain – Barré syndrome. Ann Neurol 1999; 45:168 – 73.
our opinion, fulfill this postulate and would make us less [5] Fisher M. An unusual variant of acute idiopathic polyneuritis (syn-
cautious in regard to fulfilling the postulate of antigen drome of ophthalmoplegia, ataxia and areflexia). N Engl J Med 1956;
induction of experimental disease. 225:57 – 65.
[6] The Guillain – Barré syndrome Study Group. Plasmapheresis and
(4) Disease reproduction in an experimental host by the acute Guillain – Barré syndrome. Neurology 1985;35:1096 – 104.
pathogen. No infectious animal model has yet been estab- [7] French Cooperative Group on Plasma Exchange in Guillain – Barré
lished for production of peripheral neuropathy in laboratory Syndrome. Efficiency of plasma exchange in Guillain – Barré syn-
species. A report by Li et al. [85] that 30% of chickens drome: role of replacement fluids. Ann Neurol 1987;22:753 – 61.
exposed to a C. jejuni HS:19 strain, isolated from a Chinese [8] van der Meché FGA, Schmitz PIM and the Dutch Guillain – Barré
Study Group. A randomized trial comparing intravenous globulin
patient with AMAN, developed paralysis awaits confirma- and plasma exchange in Guillain – Barré syndrome. N Engl J Med
tion. Previous studies on the structure of the glycoconjugates 1992;326:1123 – 9.
in the chicken central nervous system indicated that the [9] Illa I, Ortiz N, Gallard E, Juarez C, Grau JM, Dalakas MC. Acute
ganglioside OSs differ fundamentally from vertebrates and axonal Guillain – Barré syndrome with IgG antibody against motor
are not conserved [86] which raises similar concern for PNS axons following parenteral gangliosides. Ann Neurol 1995;38:218 –
24.
gangliosides and glycolipids. Thus, this criterion, which is [10] Nobile-Orazio E. Multifocal motor neuropathy. J Neuroimmunol
the most formidable because of species molecular differ- 2001;15:4 – 18.
ences in carbohydrates, etc., is not considered satisfied. [11] Willison HJ. Ganglioside autoantibodies. In: Peter JB, Schoenfeld Y,
However, to fulfill this requirement, it is necessary to have editors. Autoantibodies. Amsterdam, The Netherlands: Elsevier, 1996.
an animal model allowing colonization by the infectious p. 277 – 84.
[12] Moran AP, Prendergast MM. Molecular mimicry in Campylobacter
agent plus a humoral response mimicking that of humans and jejuni and Helicobacter pylori lipopolysaccharides: contribution of
found to produce a relevant pathology in animals. This is gastrointestinal infections to autoimmunity. J Autoimmun 2001;16:
proving to be difficult. 241 – 56.
In summary, there is now a substantial body of data [13] Kusunoki S, Chiba A, Kon K, Ando S, Arisawa K, Tate A, et al. N-
acetylgalactoaminyl GD1a is a target molecule for serum antibody in
indicating that there is a structural similarity of sialosyl
Guillain – Barré syndrome. Ann Neurol 1994;35:570 – 6.
determinants in gangliosides by C. jejuni LPS/LOS from [14] Ang CW, Yuki N, Jacobs BC, Koga M, van Doorn PA, Schmitz PIM,
certain and specific strains, and that this correlates signifi- et al. Rapidly progressive, predominantly motor Guillain – Barré syn-
cantly with the occurrence of GBS and related diseases. On drome with anti-GalNAc-GD1a antibodies. Neurology 1999;53:2122 –
the other hand, it is clear that other attributes of the host and/ 7.
or bacterium, in addition to molecular mimicry, are impor- [15] Yuki N, Taki T, Handa S. Antibody to GalNAc-GD1a and GalNAc-
GM1b in Guillain – Barré syndrome subsequent to Campylobacter je-
tant in the occurrence or progression of disease. These un- juni enteritis. J Neuroimmunol 1996;71:155 – 61.
determined factors may affect whether GBS develops [16] Kashihara K, Shiro Y, Koga M, Yuki N. IgG anti-GT1a antibodies
following infection with the ganglioside-mimicking strain which do not cross react with GQ1b ganglioside in a pharyngeal –
of C. jejuni. The evidence is that many the PNS disorders cervical – brachial variant of Guillain – Barré syndrome. J Neurol Neu-
initiated by C. jejuni are autoimmune, and are examples of rosurg Psychiatry, 1998. p. 65 – 799.
[17] Jacobs BC, van Doorn PA, Groeneveld JHM, Tio-Gillen AP, van der
molecular mimicry in the context of infection and induction Meché FGA. Cytomegalovirus infections and anti-GM2 antibodies in
of the neurological disorder. Guillain – Barré syndrome. J Neurol, Neurosurg Psychiatry 1997;62:
641 – 3.
[18] Khalili-Shirazi A, Gregson N, Gray IA, Rees JH, Winer JB, Hughes
RAC. Anti-ganglioside antibodies in Guillain – Barré syndrome fol-
Acknowledgements
lowing recent cytomegalovirus infection. J Neurol, Neurosurg Psy-
chiatry 1999;66:376 – 9.
These studies were supported by grants from the Irish [19] Jacobs BC, Rothbarth PH, van der Meché FGA, Herbrink P, Schmitz
Health Research Board (Grant 12-99 to A.P.M. and PI, de Klerk MA, et al. The spectrum of antecedent infections in
M.M.P.), the NIH (NS31676), and the SC Appropriation Guillain – Barré syndrome: a case-control study. Neurology 1998;51:
for Medical Research (E.L.H.). 1110 – 5.
[20] Hao Q, Saida T, Kuroki S, Nishimura M, Nukina M, Obayashi H, et
al. Antibodies to gangliosides and galactocerebroside in patients with
Guillain – Barré syndrome with preceding Campylobacter jejuni infec-
References tion and other identified infections. J Neuroimmunol 1998;81:116 –
26.
[1] Prendergast MM, Moran AP. Lipopolysaccharides in the development [21] Arnason BGW, Soliven B. Acute inflammatory demyelinating poly-
of the Guillain – Barré syndrome and Miller Fisher syndrome forms of radiculoneuropathy. In: Dyck PJ, Thomas P, Griffin JW, Low PA,
acute inflammatory peripheral neuropathies. J Endotoxin Res 2000; Poduslo J, editors. Peripheral neuropathy. Philadelphia: Saunders,
6:341 – 59. 1993. p. 1437 – 97.
[2] Willison HJ, O’Hanlon GM. The immunopathogenesis of Miller Fish- [22] Ho TW, Mishu B, Li CY, Gao Y, Cornblath DR, Griffin JW, et al. Guil-
er syndrome. J Neuroimmunol 1999;100:3 – 12. lain – Barré syndrome in northern China: relationship to Campylobacter
6 A.P. Moran et al. / Journal of the Neurological Sciences 196 (2002) 1–7

jejuni infection and anti-glycolipid antibodies. Brain 1995;118:597 – Molecular mimicry between GQ1b ganglioside and lipopolysacchar-
605. ides of Campylobacter jejuni isolated from patients with Fisher’s
[23] Allos BM, Blaser MJ. Campylobacter jejuni and the expanding spec- syndrome. Ann Neurol 1994;36:791 – 3.
trum of related infections. Clin Infect Dis 1995;20:1092 – 101. [44] Jacobs BC, Endtz HP, van der Meché FGA, Hazenberg MP, Achter-
[24] Rhodes KM, Tattersfield AK. Guillain – Barré syndrome associated eekte HAM, van Doorn PA. Serum anti-GQ1b IgG antibodies recog-
with Campylobacter infection. BMJ 1982;285:173 – 4. nize surface epitopes on Campylobacter jejuni from patients with
[25] Skirrow MB. Foodborne illness: Campylobacter. Lancet 1990;336: Miller Fisher syndrome. Ann Neurol 1995;37:260 – 4.
921 – 3. [45] Salloway S, Mermel LA, Seamens M, Aspinall GO, Nam Shin JE,
[26] Skirrow M, Fidoe RG, Jones DM. An outbreak of presumptive food- Kurjanczyk L, et al. Miller Fisher syndrome associated with Campy-
borne Campylobacter enteritis. J Infection 1981;3:234 – 6. lobacter jejuni bearing lipopolysaccharide molecules that mimic hu-
[27] Skirrow MB, Blaser MJ. Campylobacter jejuni. In: Blaser MJ, Smith man ganglioside GD3. Infect Immun 1996;64:2945 – 9.
PD, Ravdin JI, Greenberg HB, Guerrant RL, editors. Infections of the [46] Aspinall GO, McDonald AG, Pang H, Kurjanczyk LA, Penner JL.
gastrointestinal tract. New York: Raven Press, 1995. p. 825 – 48. Lipopolysaccharides of Campylobacter jejuni serotype O:19: struc-
[28] Blaser MJ, Wells JG, Feldman RA, Pollard RA, Allen JR. Campylo- tures of the core oligosaccharide regions from the serostrain and
bacter enteritis in the United States: a multicenter study. Ann Intern two bacterial isolates from patients with the Guillain – Barré syn-
Med 1983;98:360 – 5. drome. Biochemistry 1994;33:241 – 9.
[29] Moran AP, Penner JL. Serotyping of Campylobacter jejuni based on [47] Moran AP, Kosunen TU, Prendergast MM. Biochemical and serolog-
heat-stable antigens: relevance, molecular basis and implications in ical analysis of the heat-stable antigens involved in serotyping of
pathogenesis. J Appl Microbiol 1999;86:361 – 77. Campylobacter jejuni/coli. Int J Med Microbiol 2001;291(Suppl.
[30] Penner JL, Hennessy JN, Congi RV. Serotyping of Campylobacter 31):72 – 3.
jejuni and Campylobacter coli on the basis of thermostable antigens. [48] Aspinall GO, Mainkar A, Prendergast MM, Jacobs BC, Moran AP.
Eur J Clin Microbiol 1983;2:78 – 83. Lipopolysaccharides from a neuropathy-associated infection from
[31] Chart H, Frost JA, Oza A, Thwaites R, Gillanders S. Heat-stable Campylobacter jejuni serotype O:23. In: Lastovica AJ, Newell DG,
serotyping antigens expressed by strains of Campylobacter jejuni Lastovica EE, editors. Campylobacter, Helicobacter and related organ-
are probably capsular and not long-chain lipopolysaccharide. J Appl isms291. Cape Town, South Africa: Institute of Child Health and
Bacteriol 1996;81:635 – 40. University of Cape Town, 1997. p. 93 – 6.
[32] Karlyshev AV, Linton D, Gregson NA, Lastovica AJ, Wren BW. [49] Prendergast MM, Lastovica AJ, Moran AP. Lipopolysaccharides from
Genetic and biochemical evidence of a Campylobacter jejuni capsular Campylobacter jejuni O:41 strains associated with Guillain – Barré
polysaccharide that accounts for Penner serotype specificity. Mol Mi- syndrome exhibit mimicry of GM1 ganglioside. Infect Immun 1998;
crobiol 2000;35:529 – 41. 66:3649 – 55.
[33] Neisser A, Bernheimer H, Berger T, Moran AP, Schwerer B. Serum [50] Yuki N, Yoshino H, Sato S, Shinozawa K, Miyatake T. Severe acute
antibodies against gangliosides and Campylobacter jejuni lipopolysac- axonal form of Guillain – Barré syndrome associated with IgG anti-
charides in Miller Fisher syndrome. Infect Immun 1997;65: 4038 – 42. GD1a antibodies. Muscle Nerve 1992;15:899 – 903.
[34] Hanniffy OM, Shashkov AS, Moran AP, Prendergast MM, Senchen- [51] Schwerer B, Neisser A, Polt RJ, Bernheimer H, Moran AP. Antibody
kova SN, Knirel YA, et al. Chemical structure of a polysaccharide crossreactivities between gangliosides and lipopolysaccharides of
from Campylobacter jejuni 176.83 (serotype O:41) containing only Campylobacter jejuni serotypes associated with Guillain – Barré syn-
furanose sugars. Carbohydr Res 1999;319:124 – 32. drome. J Endotoxin Res 1995;2:395 – 403.
[35] Aspinall GO, Lynch CM, Pang H, Shaver RT, Moran AP. Chemical [52] Neisser A, Schwerer B, Bernheimer H, Moran AP. Ganglioside-in-
structures of the core region of Campylobacter jejuni O:3 lipopoly- duced antiganglioside antibodies from a neuropathy patient cross-react
saccharide and an associated polysaccharide. Eur J Biochem 1995; with lipopolysaccharides of Campylobacter jejuni associated with
231:570 – 8. Guillain – Barré syndrome. J Neuroimunol 2000;102:85 – 8.
[36] Aspinall GO, McDonald AG, Raju TS, Pang H, Moran AP, Penner JL. [53] Wirguin I, Suturkova-Milosevic L, Della-Latta P, Fisher T, Brown Jr
Chemical structure of the core regions of Campylobacter jejuni sero- RH, Latov N. Monoclonal IgM antibodies to GM1 and asialo-GM1 in
types O:1, O:4, O:23, and O:36 lipopolysaccharides. Eur J Biochem chronic neuropathies cross-react with Campylobacter jejuni lipopoly-
1993;213:1017 – 27. saccharides. Ann Neurol 1994;35:698 – 703.
[37] Moran AP, Rietschel ET, Kosunen TU, Zahringer U. Chemical char- [54] Prendergast MM, Willison HJ, Moran AP. Human monoclonal IgM
acterization of Campylobacter jejuni lipopolysaccharides containing antibodies to GM1 ganglioside show diverse cross-reactivity with lip-
N-acetylneuraminic acid and 2, 3-diamino-2, 3-dideoxy-D-glucose. J opolysaccharides of Campylobacter jejuni strains associated with
Bacteriol 1991;173:618 – 26. Guillain – Barré syndrome. Infect Immun 1999;67:3698 – 701.
[38] Kuroki S, Saida T, Nukina M, Haruta T, Yoshiota M, Kobayashi Y, et [55] Goodyear CS, O’Hanlon GM, Plomp JJ, Wagner ER, Morrison I,
al. Campylobacter jejuni strains from patients with Guillain – Barré Veitch J, et al. Monoclonal antibodies raised against Guillain – Barré
syndrome belong mostly to Penner serogroup 19 and contain h-N- syndrome-associated Campylobacter jejuni lipopolysaccharides react
acetylglucosamine residues. Ann Neurol 1993;33:243 – 7. with neuronal gangliosides and paralyze muscle – nerve preparations. J
[39] Fujimoto S, Yuki N, Itoh T, Amako K. Specific serotype of Campy- Clin Invest 1999;104:697 – 708.
lobacter jejuni associated with Guillain – Barré syndrome. J Infect Dis [56] Prendergast MM, Moran AP. Antiganglioside antibodies induced by
1992;165:183. ganglioside-mimicking Campylobacter jejuni lipopolysaccharide bind
[40] Moran AP, O’Malley DT. Potential role of lipopolysaccharides of to epitopes at the nodes of Ranvier. Int J Med Microbiol 2001;291
Campylobacter jejuni in the development of Guillain – Barré syn- (Suppl. 31):125.
drome. J Endotoxin Res 1995;2:233 – 5. [57] The Dutch Guillain – Barré Study Group. Treatment of Guillain – Barré
[41] Mishu B, Ilyas AA, Koski CL, Vriesendorp FJ, Cook SD, Milton FA, syndrome with high-dose immune globulins combined with methyl-
et al. Serologic evidence of previous Campylobacter jejuni infection prednisolone: a pilot study. Ann Neurol 1994;35:749 – 52.
in patients with the Guillain – Barré syndrome. Ann Intern Med 1993; [58] Plasma Exchange/Sandoglobulin Guillain – Barré Syndrome Trial
118:947 – 57. Group. Randomised trial of plasma exchange, intravenous immuno-
[42] Lastovica AJ, Goddard EA, Argent AC. Guillain – Barré syndrome in globulin, and combined treatments in Guillain – Barré syndrome. Lan-
South Africa associated with Campylobacter jejuni O:41 strains. J cet 1997;349:225 – 30.
Infect Dis 1997;176(Suppl. 2):139 – 43. [59] Witebsky E, Rose NR, Terplan K, Pairie JR, Egan RW. Chronic thyroi-
[43] Yuki N, Taki T, Takihashi M, Saito K, Yoshino H, Tai T, et al. ditis and autoimmunity. J Am Med Assoc 1957;164:1439 – 47.
A.P. Moran et al. / Journal of the Neurological Sciences 196 (2002) 1–7 7

[60] Sheikh KA, Griffin JW. Variants of the Guillain – Barré syndrome: al. IgM deposits at the nodes of Ranvier in a patient with amytrophic
progress toward fulfilling ‘‘Koch’s postulates’’. Ann Neurol 2001; lateral sclerosis, anti-GM1 antibodies, and multifocal motor conduc-
49:694 – 6. tion block. Ann Neurol 1990;28:373 – 7.
[61] Prendergast MM, Kosunen TU, Moran AP. Development of an im- [74] Sumner A, Said G, Idy I, Metral S. Demyelinative conduction block
munoassay for rapid detection of ganglioside GM1 mimicry in Cam- produced by intraneural injection of human Guillain – Barré serum
pylobacter jejuni strains. J Clin Microbiol 2001;39:1494 – 500. into rat sciatic nerve. Neurology 1982;32:106.
[62] Oomes PG, Jacobs BC, Hazenberg MPH, Bänffer JRJ, van der Meché [75] Feasby TE, Hahn AF, Gilbert JJ. Passive transfer studies in Guillain –
FGA. Anti-GM1 IgG antibodies and Campylobacter bacteria in Guil- Barré polyneuropathy. Neurology 1982;32:1159 – 67.
lain – Barré syndrome: evidence of molecular mimicry. Ann Neurol [76] Saida T, Saida K, Lisak RP, Brown MJ, Silberberg DH, Asbury AK.
1995;38:170 – 5. In vivo demyelinating activity of sera from patients with Guillain –
[63] Sheikh KA, Nachamkin I, Ho T, Willison HJ, Veitch J, Ung H, et al. Barré syndrome. Ann Neurol 1982;11:69 – 75.
Campylobacter jejuni lipopolysaccharide in Guillain – Barré syndrome: [77] Harrison BM, Hansen LA, Pollard JD, McLeod JG. Demyelination
molecular mimicry and host susceptibility. Neurology 1998;51:371 – 8. induced by serum from patients with Guillain – Barré syndrome. Ann
[64] Paparounas K, O’Hanlon GM, O’Leary CP, Rowan EG, Willison HJ. Neurol 1984;15:163 – 70.
Anti-ganglioside antibodies can bind peripheral nerve nodes of Ranv- [78] Hafer-Macko C, Hsieh ST, Li CY, Ho TW, Sheihk K, Cornblath DR,
ier and activate the complement cascade without inducing acute con- et al. Acute motor axonal neuropathy; an antibody-mediated attack on
duction block in vitro. Brain 1999;122:807 – 16. axolemma. Ann Neurol 1996;40:635 – 44.
[65] O’Hanlon GM, Paterson GJ, Veitch J, Wilson G, Willison HJ. Map- [79] Hartung HP, Toyka KV, Griffin JW. Guillain – Barré syndrome and
ping immunoreactive epitopes in the human peripheral nervous sys- chronic idiopathic demyelinating polyneuropathy. In: Antel J, Birn-
tem using human monoclonal anti-GM1 ganglioside antibodies. Acta baum G, Hartung HP, editors. Clinical neuroimmunology. Oxford,
Neuropathol (Berlin) 1998;95:605 – 16. UK: Blackwell, 1998. p. 294 – 306.
[66] Corbo M, Quattrini A, Latov N, Hays AP. Localisation of GM1 and [80] Buchwald B, Bufler J, Carpo M, et al. Combined pre- and postsynap-
Gal(h1 – 3) GalNAc antigenic determinants in peripheral nerve. Neu- tic action of IgG antibodies in Miller Fisher syndrome. Neurology
rology 1993;43:809 – 14. 2001;56:67 – 74.
[67] Plomp JJ, Molenaar PC, O’Hanlon GM, Jacobs BC, Veitch J, Daha [81] Yuki N, Yamada M, Koga M, Odaka M, Susuki K, Tagawa Y, et al.
MR, et al. Miller Fisher anti-GQ1b antibodies: alpha-latrotoxin-like Animal model of axonal Guillain – Barre syndrome induced by sensi-
effects on motor end plates. Ann Neurol 1999;45:189 – 99. tization with GM1 ganglioside. Ann Neurol 2001;49:712 – 20.
[68] Thomas FP, Adapon PH, Goldberg GP, Latov N, Hays AP. Local- [82] Buchwald B, Toyka KV, Zielasek J, Weishaupt A, Schweiger S, Dudel
isation of neural epitopes that bind to IgM monoclonal autoantibodies J. Neuromuscular blockade by IgG antibodies from patients with
(M-proteins) from two patients with motor neuron disease. J Neuro- Guillain – Barre syndrome; a macro-patch-clamp study. Ann Neurol
immunol 1989;21:31 – 9. 1998;44:913 – 22.
[69] Araski K, Kusunoki S, Kudo M, Tamaki M. The pattern of antigan- [83] Moran AP, Prendergast MM. Molecular mimicry in Campylobacter
glioside antibody reactivities producing myelinated nerve conduction jejuni lipopolysaccharides and the development of Guillain – Barré
block in vitro. J Neurol Sci 1998;161:163 – 8. syndrome. J Infect Dis 1998;178:1549 – 50.
[70] Takigawa T, Yasuda H, Kikkawa R, Shigeta Y, Saida T, Kitasata H. [84] Kusunoki S, Hitoshi S, Kaida K, Murayama S, Kanazawa I. Degen-
Antibodies against GM1 affect K + and Na + currents in isolated rat eration of rabbit sensory neurons induced by passive transfer of anti-
myelinated nerve fibres. Ann Neurol 1995;37:436 – 42. GD1b antiserum. Neurosci Lett 1999;273:33 – 6.
[71] Roberts M, Willison H, Vincent A, Newsome-Davis J. Serum factor in [85] Li CY, Xue P, Tian WQ, Liu RC, Yang C. Experimental Campylo-
miller Fisher variant of Guillain – Barré syndrome and neurotransmit- bacter jejuni infection in the chicken: an animal model of axonal
ter release. Lancet 1994;343:454 – 5. Guillain – Barré syndrome. J Neurol, Neurosurg Psychiatry 1996; 61:
[72] Roberts M, Willison H, Vincent A, Newsome-Davis J. Multifocal 279 – 84.
motor neuropathy human sera block distal nerve conduction in mice. [86] Chien JL, Hogan EL. Glycosphingolipids of skeletal muscle. In:
Ann Neurol 1995;38:111 – 8. Sweeley CC, editor. Cell surface glycolipids. ACS Symp Ser, vol.
[73] Santoro M, Thomas FP, Fink ME, Lange DJ, Uncini A, Wadia NH, et 128. Washington D.C., USA: Am Chem Soc; 1980. p. 135 – 48.

You might also like