Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Peripher Nerv Syst ; 28(1): 32-40, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36573790

RESUMO

Recent work identified anti-GM2 and anti-GalNAc-GD1a IgG ganglioside antibodies as biomarkers in dogs clinically diagnosed with acute canine polyradiculoneuritis, in turn considered a canine equivalent of Guillain-Barré syndrome. This study aims to investigate the serum prevalence of similar antibodies in cats clinically diagnosed with immune-mediated polyneuropathies. The sera from 41 cats clinically diagnosed with immune-mediated polyneuropathies (IPN), 9 cats with other neurological or neuromuscular disorders (ONM) and 46 neurologically normal cats (CTRL) were examined for the presence of IgG antibodies against glycolipids GM1, GM2, GD1a, GD1b, GalNAc-GD1a, GA1, SGPG, LM1, galactocerebroside and sulphatide. A total of 29/41 IPN-cats had either anti-GM2 or anti-GalNAc-GD1a IgG antibodies, with 24/29 cats having both. Direct comparison of anti-GM2 (sensitivity: 70.7%; specificity: 78.2%) and anti-GalNAc-GD1a (sensitivity: 70.7%; specificity: 70.9%) antibodies narrowly showed anti-GM2 IgG antibodies to be the better marker for identifying IPN-cats when compared to the combined ONM and CTRL groups (P = .049). Anti-GA1 and/or anti-sulphatide IgG antibodies were ubiquitously present across all sample groups, whereas antibodies against GM1, GD1a, GD1b, SGPG, LM1 and galactocerebroside were overall only rarely observed. Anti-GM2 and anti-GalNAc-GD1a IgG antibodies may serve as serum biomarkers for immune-mediated polyneuropathies in cats, as previously observed in dogs and humans.


Assuntos
Síndrome de Guillain-Barré , Polineuropatias , Humanos , Gatos , Animais , Cães , Galactosilceramidas , Gangliosídeo G(M1) , Gangliosídeos , Imunoglobulina G , Polineuropatias/diagnóstico , Polineuropatias/veterinária , Biomarcadores , Autoanticorpos , Gangliosídeo G(M2)
2.
Lancet ; 387(10027): 1531-1539, 2016 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-26948433

RESUMO

BACKGROUND: Between October, 2013, and April, 2014, French Polynesia experienced the largest Zika virus outbreak ever described at that time. During the same period, an increase in Guillain-Barré syndrome was reported, suggesting a possible association between Zika virus and Guillain-Barré syndrome. We aimed to assess the role of Zika virus and dengue virus infection in developing Guillain-Barré syndrome. METHODS: In this case-control study, cases were patients with Guillain-Barré syndrome diagnosed at the Centre Hospitalier de Polynésie Française (Papeete, Tahiti, French Polynesia) during the outbreak period. Controls were age-matched, sex-matched, and residence-matched patients who presented at the hospital with a non-febrile illness (control group 1; n=98) and age-matched patients with acute Zika virus disease and no neurological symptoms (control group 2; n=70). Virological investigations included RT-PCR for Zika virus, and both microsphere immunofluorescent and seroneutralisation assays for Zika virus and dengue virus. Anti-glycolipid reactivity was studied in patients with Guillain-Barré syndrome using both ELISA and combinatorial microarrays. FINDINGS: 42 patients were diagnosed with Guillain-Barré syndrome during the study period. 41 (98%) patients with Guillain-Barré syndrome had anti-Zika virus IgM or IgG, and all (100%) had neutralising antibodies against Zika virus compared with 54 (56%) of 98 in control group 1 (p<0.0001). 39 (93%) patients with Guillain-Barré syndrome had Zika virus IgM and 37 (88%) had experienced a transient illness in a median of 6 days (IQR 4-10) before the onset of neurological symptoms, suggesting recent Zika virus infection. Patients with Guillain-Barré syndrome had electrophysiological findings compatible with acute motor axonal neuropathy (AMAN) type, and had rapid evolution of disease (median duration of the installation and plateau phases was 6 [IQR 4-9] and 4 days [3-10], respectively). 12 (29%) patients required respiratory assistance. No patients died. Anti-glycolipid antibody activity was found in 13 (31%) patients, and notably against GA1 in eight (19%) patients, by ELISA and 19 (46%) of 41 by glycoarray at admission. The typical AMAN-associated anti-ganglioside antibodies were rarely present. Past dengue virus history did not differ significantly between patients with Guillain-Barré syndrome and those in the two control groups (95%, 89%, and 83%, respectively). INTERPRETATION: This is the first study providing evidence for Zika virus infection causing Guillain-Barré syndrome. Because Zika virus is spreading rapidly across the Americas, at risk countries need to prepare for adequate intensive care beds capacity to manage patients with Guillain-Barré syndrome. FUNDING: Labex Integrative Biology of Emerging Infectious Diseases, EU 7th framework program PREDEMICS. and Wellcome Trust.


Assuntos
Surtos de Doenças , Síndrome de Guillain-Barré/epidemiologia , Síndrome de Guillain-Barré/virologia , Infecção por Zika virus/complicações , Infecção por Zika virus/epidemiologia , Adulto , Estudos de Casos e Controles , Vírus da Dengue/isolamento & purificação , Surtos de Doenças/estatística & dados numéricos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Polinésia/epidemiologia , Dengue Grave/complicações , Dengue Grave/epidemiologia , Zika virus/isolamento & purificação
3.
J Peripher Nerv Syst ; 22(1): 4-12, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27801990

RESUMO

The outcome of Guillain-Barré syndrome (GBS) remains unchanged since plasma exchange and intravenous immunoglobulin (IVIg) were introduced over 20 years ago. Pathogenesis studies on GBS have identified the terminal component of complement cascade as a key disease mediator and therapeutic target. We report the first use of terminal complement pathway inhibition with eculizumab in humans with GBS. In a randomised, double-blind, placebo-controlled trial, 28 subjects eligible on the basis of GBS disability grade of at least 3 were screened, of whom 8 (29%) were randomised. Five received eculizumab for 4 weeks, alongside standard IVIg treatment. The safety outcomes, monitored via adverse events capture, showed eculizumab to be well-tolerated and safe when administered in conjunction with IVIg. Primary and secondary efficacy outcomes in the form of GBS disability scores (GBS DS), MRC sum scores, Rasch overall disability scores, and overall neuropathy limitation scores are reported descriptively. For the primary efficacy outcome at 4 weeks after recruitment, two of two placebo- and two of five eculizumab-treated subjects had improved by one or more grades on the GBS DS. Although the small sample size precludes a statistically meaningful analysis, these pilot data indicate further studies on complement inhibition in GBS are warranted.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome de Guillain-Barré/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Adulto , Idoso , Avaliação da Deficiência , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Gangliosidoses GM2/metabolismo , Gangliosidose GM1/metabolismo , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
4.
Brain ; 139(Pt 6): 1657-65, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27017187

RESUMO

SEE VAN DOORN AND JACOBS DOI101093/BRAIN/AWW078 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE : In axonal forms of Guillain-Barré syndrome, anti-ganglioside antibodies bind gangliosides on nerve surfaces, thereby causing injury through complement activation and immune cell recruitment. Why some nerve regions are more vulnerable than others is unknown. One reason may be that neuronal membranes with high endocytic activity, including nerve terminals involved in neurotransmitter recycling, are able to endocytose anti-ganglioside antibodies from the cell surface so rapidly that antibody-mediated injury is attenuated. Herein we investigated whether endocytic clearance of anti-ganglioside antibodies by nerve terminals might also be of sufficient magnitude to deplete circulating antibody levels. Remarkably, systemically delivered anti-ganglioside antibody in mice was so avidly cleared from the circulation by endocytosis at ganglioside-expressing plasma membranes that it was rapidly rendered undetectable in serum. A major component of the clearance occurred at motor nerve terminals of neuromuscular junctions, from where anti-ganglioside antibody was retrogradely transported to the motor neuron cell body in the spinal cord, recycled to the plasma membrane, and secreted into the surrounding spinal cord. Uptake at the neuromuscular junction represents a major unexpected pathway by which pathogenic anti-ganglioside antibodies, and potentially other ganglioside binding proteins, are cleared from the systemic circulation and also covertly delivered to the central nervous system.


Assuntos
Anticorpos/metabolismo , Endocitose/imunologia , Gangliosídeos/imunologia , Terminações Pré-Sinápticas/metabolismo , Animais , Anticorpos/sangue , Membrana Celular/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Knockout , N-Acetilgalactosaminiltransferases/genética , Junção Neuromuscular/metabolismo , Polipeptídeo N-Acetilgalactosaminiltransferase
5.
Artigo em Inglês | MEDLINE | ID: mdl-36411078

RESUMO

BACKGROUND AND OBJECTIVES: Recent outbreaks of Zika virus (ZIKV) in South and Central America have highlighted significant neurologic side effects. Concurrence with the inflammatory neuropathy Guillain-Barré syndrome (GBS) is observed in 1:4,000 ZIKV cases. Whether the neurologic symptoms of ZIKV infection are immune mediated is unclear. We used rodent and human live cellular models to screen for anti-peripheral nerve reactive IgG and IgM autoantibodies in the sera of patients with ZIKV with and without GBS. METHODS: In this study, 52 patients with ZIKV-GBS were compared with 134 ZIKV-infected patients without GBS and 91 non-ZIKV controls. Positive sera were taken forward for target identification by immunoprecipitation and mass spectrometry, and candidate antigens were validated by ELISA and cell-based assays. Autoantibody reactions against glycolipid antigens were also screened on an array. RESULTS: Overall, IgG antibody reactivities to rat Schwann cells (SCs) (6.5%) and myelinated cocultures (9.6%) were significantly higher, albeit infrequent, in the ZIKV-GBS group compared with all controls. IgM antibody immunoreactivity to dorsal root ganglia neurones (32.3%) and SCs (19.4%) was more frequently observed in the ZIKV-GBS group compared with other controls, whereas IgM reactivity to cocultures was as common in ZIKV and non-ZIKV sera. Strong axonal-binding ZIKV-GBS serum IgG antibodies from 1 patient were confirmed to react with neurofascin 155 and 186. Serum from a ZIKV-infected patient without GBS displayed strong myelin-binding and putative antilipid antigen reaction characteristics. There was, however, no significant association of ZIKV-GBS with any known antiglycolipid antibodies. DISCUSSION: Autoantibody responses in ZIKV-GBS target heterogeneous peripheral nerve antigens suggesting heterogeneity of the humoral immune response despite a common prodromal infection.


Assuntos
Síndrome de Guillain-Barré , Infecção por Zika virus , Zika virus , Humanos , Animais , Ratos , Infecção por Zika virus/complicações , Infecção por Zika virus/epidemiologia , Síndrome de Guillain-Barré/diagnóstico , Imunoglobulina M , Imunoglobulina G , Autoanticorpos
6.
Methods Mol Biol ; 2460: 183-191, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34972937

RESUMO

Glycolipids cluster in plasma membranes to form heterogenous patches. Whereas lectins and antibodies have been conventionally viewed as binding a single oligosaccharide head group, and assayed accordingly, it is increasingly evident that cis-interactions between glycan headgroups may form unique molecular shapes that either enhance or attenuate binding of antibodies and other proteins. Herein we describe a method for assaying antibody binding to heteromeric glycolipid complexes that allows rapid, simple, inexpensive and high-throughput assessment of binding events, focusing on autoantibodies present in human serum.


Assuntos
Autoanticorpos , Glicolipídeos , Gangliosídeos , Glicolipídeos/química , Humanos , Lectinas/metabolismo , Análise em Microsséries/métodos , Polissacarídeos
7.
Brain ; 133(Pt 7): 1944-60, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20513658

RESUMO

The motor axonal variant of Guillain-Barré syndrome is associated with anti-GD1a immunoglobulin antibodies, which are believed to be the pathogenic factor. In previous studies we have demonstrated the motor terminal to be a vulnerable site. Here we show both in vivo and ex vivo, that nodes of Ranvier in intramuscular motor nerve bundles are also targeted by anti-GD1a antibody in a gradient-dependent manner, with greatest vulnerability at distal nodes. Complement deposition is associated with prominent nodal injury as monitored with electrophysiological recordings and fluorescence microscopy. Complete loss of nodal protein staining, including voltage-gated sodium channels and ankyrin G, occurs and is completely protected by both complement and calpain inhibition, although the latter provides no protection against electrophysiological dysfunction. In ex vivo motor and sensory nerve trunk preparations, antibody deposits are only observed in experimentally desheathed nerves, which are thereby rendered susceptible to complement-dependent morphological disruption, nodal protein loss and reduced electrical activity of the axon. These studies provide a detailed mechanism by which loss of axonal conduction can occur in a distal dominant pattern as observed in a proportion of patients with motor axonal Guillain-Barré syndrome, and also provide an explanation for the occurrence of rapid recovery from complete paralysis and electrophysiological in-excitability. The study also identifies therapeutic approaches in which nodal architecture can be preserved.


Assuntos
Autoanticorpos/toxicidade , Calpaína/metabolismo , Ativação do Complemento/imunologia , Gangliosídeos/imunologia , Neurônios Motores/imunologia , Neurônios Motores/patologia , Nós Neurofibrosos/imunologia , Nós Neurofibrosos/patologia , Animais , Autoanticorpos/metabolismo , Axônios/imunologia , Axônios/patologia , Sítios de Ligação de Anticorpos , Síndrome de Guillain-Barré/imunologia , Síndrome de Guillain-Barré/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Neurônios Motores/metabolismo , Nós Neurofibrosos/metabolismo
8.
Artigo em Inglês | MEDLINE | ID: mdl-33547152

RESUMO

OBJECTIVE: To identify the clinical phenotypes and infectious triggers in the 2019 Peruvian Guillain-Barré syndrome (GBS) outbreak. METHODS: We prospectively collected clinical and neurophysiologic data of patients with GBS admitted to a tertiary hospital in Lima, Peru, between May and August 2019. Molecular, immunologic, and microbiological methods were used to identify causative infectious agents. Sera from 41 controls were compared with cases for antibodies to Campylobacter jejuni and gangliosides. Genomic analysis was performed on 4 C jejuni isolates. RESULTS: The 49 included patients had a median age of 44 years (interquartile range [IQR] 30-54 years), and 28 (57%) were male. Thirty-two (65%) had symptoms of a preceding infection: 24 (49%) diarrhea and 13 (27%) upper respiratory tract infection. The median time between infectious to neurologic symptoms was 3 days (IQR 2-9 days). Eighty percent had a pure motor form of GBS, 21 (43%) had the axonal electrophysiologic subtype, and 18% the demyelinating subtype. Evidence of recent C jejuni infection was found in 28/43 (65%). No evidence of recent arbovirus infection was found. Twenty-three cases vs 11 controls (OR 3.3, confidence interval [CI] 95% 1.2-9.2, p < 0.01) had IgM and/or IgA antibodies against C jejuni. Anti-GM1:phosphatidylserine and/or anti-GT1a:GM1 heteromeric complex antibodies were strongly positive in cases (92.9% sensitivity and 68.3% specificity). Genomic analysis showed that the C jejuni strains were closely related and had the Asn51 polymorphism at cstII gene. CONCLUSIONS: Our study indicates that the 2019 Peruvian GBS outbreak was associated with C jejuni infection and that the C jejuni strains linked to GBS circulate widely in different parts of the world.


Assuntos
Infecções por Campylobacter/diagnóstico , Infecções por Campylobacter/epidemiologia , Campylobacter jejuni/isolamento & purificação , Surtos de Doenças , Síndrome de Guillain-Barré/diagnóstico , Síndrome de Guillain-Barré/epidemiologia , Adulto , Infecções por Campylobacter/sangue , Estudos de Casos e Controles , Feminino , Síndrome de Guillain-Barré/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Peru/epidemiologia
10.
Brain ; 131(Pt 5): 1197-208, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18184663

RESUMO

Anti-GQ1b ganglioside antibodies are the serological hallmark of the Miller Fisher syndrome (MFS) variant of the paralytic neuropathy, Guillain-Barré syndrome, and are believed to be the principal pathogenic mediators of the disease. In support of this, we previously showed in an in vitro mouse model of MFS that anti-GQ1b antibodies were able to bind and disrupt presynaptic motor nerve terminals at the neuromuscular junction (NMJ) as one of their target sites, thereby causing muscle paralysis. This injury only occurred through activation of complement, culminating in the formation and deposition of membrane attack complex (MAC, C5b-9) in nerve membranes. Since this step is crucial to the neuropathic process and an important convergence point for antibody and complement mediated membrane injury in general, it forms an attractive pharmacotherapeutic target. Here, we assessed the efficacy of the humanized monoclonal antibody eculizumab, which blocks the formation of human C5a and C5b-9, in preventing the immune-mediated motor neuropathy exemplified in this model. Eculizumab completely prevented electrophysiological and structural lesions at anti-GQ1b antibody pre-incubated NMJs in vitro when using normal human serum (NHS) as a complement source. In a novel in vivo mouse model of MFS generated through intraperitoneal injection of anti-GQ1b antibody and NHS, mice developed respiratory paralysis due to transmission block at diaphragm NMJs, resulting from anti-GQ1b antibody binding and complement activation. Intravenous injection of eculizumab effectively prevented respiratory paralysis and associated functional and morphological hallmarks of terminal motor neuropathy. We show that eculizumab protects against complement-mediated damage in murine MFS, providing the rationale for undertaking clinical trials in this disease and other antibody-mediated neuropathies in which complement activation is believed to be involved.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Síndrome de Miller Fisher/prevenção & controle , Doença Autoimune do Sistema Nervoso Experimental/prevenção & controle , Animais , Anticorpos Monoclonais Humanizados , Ativação do Complemento/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Gangliosídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Síndrome de Miller Fisher/imunologia , Síndrome de Miller Fisher/fisiopatologia , Contração Muscular , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Doença Autoimune do Sistema Nervoso Experimental/fisiopatologia , Junção Neuromuscular/imunologia , Junção Neuromuscular/fisiopatologia , Paralisia Respiratória/imunologia , Paralisia Respiratória/fisiopatologia , Paralisia Respiratória/prevenção & controle , Sinapses/ultraestrutura , Técnicas de Cultura de Tecidos
11.
J Neuroimmunol ; 201-202: 172-82, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18664403

RESUMO

Recent research into the Guillain-Barré syndromes (GBS) has focused on anti-ganglioside antibodies that correlate with specific clinical phenotypes. Our increasing understanding of the role of antibodies in mediating GBS has naturally focused our attention on complement involvement in the pathological procession. We have studied the axonal and glial components of the murine motor nerve terminal as a model site of antibody and complement mediated injury. Such studies are providing us with clear information on the molecular components underlying our clinicopathological model for GBS and have lead us to the testing of emerging complement therapeutics that are potentially suitable for human use.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Síndrome de Guillain-Barré/complicações , Fatores Imunológicos/fisiologia , Doenças do Sistema Nervoso Periférico/etiologia , Doenças do Sistema Nervoso Periférico/patologia , Animais , Anticorpos/metabolismo , Modelos Animais de Doenças , Gangliosídeos/imunologia , Síndrome de Guillain-Barré/tratamento farmacológico , Humanos , Fatores Imunológicos/farmacologia , Camundongos , Modelos Biológicos , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/metabolismo , Doenças do Sistema Nervoso Periférico/tratamento farmacológico
12.
J Peripher Nerv Syst ; 13(3): 228-35, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18844789

RESUMO

Guillain-Barré syndrome and its clinical variants, including the anti-GQ1b ganglioside-mediated Miller Fisher syndrome (MFS), comprise the world's leading cause of acute neuromuscular paralysis. Presently, no specific drug therapies exist. The complement cascade, which is activated in these patients, forms an attractive drug target. In this study, we tested whether the complement C5-inhibiting recombinant protein, rEV576, was able to prevent neural injury in a previously developed in vitro mouse model for MFS. Mouse hemidiaphragm preparations were treated with anti-GQ1b antibody and normal human serum as a source of complement with added rEV576 or control protein. Immunohistology in control tissue showed deposition of C3c and membrane attack complex at neuromuscular junctions (NMJs), along with terminal motor axonal neurofilament degradation as well as ethidium homodimer-2 staining showing perisynaptic Schwann cell (pSC) injury. Electrophysiological and functional analyses showed block of synaptic transmission at the NMJ after an initial period of a dramatically high level of asynchronous acetylcholine release. In tissue treated with rEV576, all these indicators of motor neuronal damage were absent, except for the presence of C3c, indicating effective inhibition of C5. These results demonstrate that rEV576 effectively prevents development of neuronal and pSC damage in experimental murine neuropathy.


Assuntos
Complemento C5/antagonistas & inibidores , Proteínas Inativadoras do Complemento/farmacologia , Complexo de Ataque à Membrana do Sistema Complemento/efeitos dos fármacos , Síndrome de Miller Fisher/tratamento farmacológico , Junção Neuromuscular/efeitos dos fármacos , Animais , Diafragma/efeitos dos fármacos , Modelos Animais de Doenças , Potenciais Evocados/efeitos dos fármacos , Imunofluorescência , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Proteínas de Insetos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Técnicas de Cultura de Órgãos , Proteínas Recombinantes/farmacologia
13.
J Neuroimmunol ; 323: 28-35, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30196830

RESUMO

Sulfatide is a major glycosphingolipid in myelin and a target for autoantibodies in autoimmune neuropathies. However neuropathy disease models have not been widely established, in part because currently available monoclonal antibodies to sulfatide may not represent the diversity of anti-sulfatide antibody binding patterns found in neuropathy patients. We sought to address this issue by generating and characterising a panel of new anti-sulfatide monoclonal antibodies. These antibodies have sulfatide reactivity distinct from existing antibodies in assays and in binding to peripheral nerve tissues and can be used to provide insights into the pathophysiological roles of anti-sulfatide antibodies in demyelinating neuropathies.


Assuntos
Autoanticorpos/sangue , Membrana Celular/metabolismo , Neuroglia/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/patologia , Células Cultivadas , Doenças Desmielinizantes/sangue , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Neuroglia/patologia , Ligação Proteica/fisiologia
14.
J Neurosci ; 25(7): 1620-8, 2005 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-15716397

RESUMO

Anti-GD1a ganglioside antibodies (Abs) are the serological hallmark of the acute motor axonal form of the post-infectious paralysis, Guillain-Barre syndrome. Development of a disease model in mice has been impeded by the weak immunogenicity of gangliosides and the apparent resistance of GD1a-containing neural membranes to anti-GD1a antibody-mediated injury. Here we used mice with altered ganglioside biosynthesis to generate such a model at motor nerve terminals. First, we bypassed immunological tolerance by immunizing GD1a-deficient, beta-1,4-N-acetylgalactosaminyl transferase knock-out mice with GD1a ganglioside-mimicking antigens from Campylobacter jejuni and generated high-titer anti-GD1a antisera and complement fixing monoclonal Abs (mAbs). Next, we exposed ex vivo nerve-muscle preparations from GD1a-overexpressing, GD3 synthase knock-out mice to the anti-GD1a mAbs in the presence of a source of complement and investigated morphological and electrophysiological damage. Dense antibody and complement deposits were observed only over presynaptic motor axons, accompanied by severe ultrastructural damage and electrophysiological blockade of motor nerve terminal function. Perisynaptic Schwann cells and postsynaptic membranes were unaffected. In contrast, normal mice were not only unresponsive to immunization with GD1a but also resistant to neural injury during anti-GD1a Ab exposure, demonstrating the central role of membrane antigen density in modulating both immune tolerance to GD1a and axonal susceptibility to anti-GD1a Abmediated injury. Identical paralyzing effects were observed when testing mouse and human anti-GD1a-positive sera. These data indicate that anti-GD1a Abs arise via molecular mimicry and are likely to be clinically relevant in injuring peripheral nerve axonal membranes containing sufficiently high levels of GD1a.


Assuntos
Autoanticorpos/imunologia , Autoantígenos/imunologia , Modelos Animais de Doenças , Gangliosídeos/imunologia , Mimetismo Molecular/imunologia , Neurônios Motores/fisiologia , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Junção Neuromuscular/imunologia , Animais , Antígenos de Bactérias/imunologia , Autoantígenos/biossíntese , Axônios/imunologia , Campylobacter jejuni/imunologia , Ativação do Complemento , Gangliosídeos/biossíntese , Síndrome de Guillain-Barré/etiologia , Síndrome de Guillain-Barré/imunologia , Tolerância Imunológica , Imunização , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Neurônios Motores/imunologia , N-Acetilgalactosaminiltransferases/deficiência , N-Acetilgalactosaminiltransferases/genética , Doença Autoimune do Sistema Nervoso Experimental/etiologia , Junção Neuromuscular/fisiopatologia , Junção Neuromuscular/ultraestrutura , Sialiltransferases/deficiência , Sialiltransferases/genética , Polipeptídeo N-Acetilgalactosaminiltransferase
15.
Neurol Neuroimmunol Neuroinflamm ; 3(6): e284, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27790627

RESUMO

OBJECTIVE: To characterize the patterns of autoantibodies to glycolipid complexes in a large cohort of Guillain-Barré syndrome (GBS) and control samples collected in Bangladesh using a newly developed microarray technique. METHODS: Twelve commonly studied glycolipids and lipids, plus their 66 possible heteromeric complexes, totaling 78 antigens, were applied to polyvinylidene fluoride-coated slides using a microarray printer. Arrays were probed with 266 GBS and 579 control sera (2 µL per serum, diluted 1/50) and bound immunoglobulin G detected with secondary antibody. Scanned arrays were subjected to statistical analyses. RESULTS: Measuring antibodies to single targets was 9% less sensitive than to heteromeric complex targets (49.2% vs 58.3%) without significantly affecting specificity (83.9%-85.0%). The optimal screening protocol for GBS sera comprised a panel of 10 glycolipids (4 single glycolipids GM1, GA1, GD1a, GQ1b, and their 6 heteromeric complexes), resulting in an overall assay sensitivity of 64.3% and specificity of 77.1%. Notable heteromeric targets were GM1:GD1a, GM1:GQ1b, and GA1:GD1a, in which exclusive binding to the complex was observed. CONCLUSIONS: Rationalizing the screening protocol to capture the enormous diversity of glycolipid complexes can be achieved by miniaturizing the screening platform to a microarray platform, and applying simple bioinformatics to determine optimal sensitivity and specificity of the targets. Glycolipid complexes are an important category of glycolipid antigens in autoimmune neuropathy cases that require specific analytical and bioinformatics methods for optimal detection.

16.
Brain ; 127(Pt 9): 2109-23, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15289269

RESUMO

Anti-disialoside antibodies (Abs) that bind NeuAc(alpha2-8) NeuAc epitopes on GQ1b and related gangliosides are found in human autoimmune neuropathy sera and are considered to be pathogenic. In a model system in mice, one mechanism by which anti-disialoside Abs have been demonstrated to induce paralysis is through a complement dependent blocking effect on transmitter release at the neuromuscular junction, similar to the effects of alpha-latrotoxin. Although direct targeting of presynaptic neuronal membranes occurs in this model, concomitant injury to perisynaptic Schwann cells (pSC) could indirectly contribute to this paralytic effect by influencing nerve terminal function and survival. To examine this possibility and the specific complement components that might mediate these effects, we exposed neuromuscular junctions in vivo and in vitro to an anti-disialoside Ab in conjunction with intact and selectively deficient complement sources. Using immuno-electron microscopy, we observed Ab deposits equally distributed on both neuronal and pSC membranes, and ultrastructural evidence of injury at both sites. Presynaptic neuronal injury was demonstrated functionally with microelectrode recordings and histologically as neurofilament loss. As hypothesized, concomitant pSC injury occurred, as indicated by abnormal uptake of ethidium dimer into pSC nuclei. The pSC and nerve terminal damage indicators correlated well with deposition of the pore-forming terminal complement component, membrane attack complex (MAC) in pSC and nerve terminal membranes. Furthermore, both neuronal and pSC injury were exacerbated in tissues from mice lacking the inhibitory complement regulator, CD59, where MAC formation is increased. These data demonstrate that both presynaptic neuronal membranes and pSCs are targets for anti-disialoside Abs, and that the injury to both sites is mediated by MAC and further regulated by CD59. This is the first demonstration that complement mediated pSC injury occurs in a model of autoimmune neuropathy and provides a rationale for investigating the possibility of pSC injury in equivalent conditions in man.


Assuntos
Anticorpos/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Gangliosídeos/imunologia , Neurônios Motores/imunologia , Fatores de Crescimento Neural/imunologia , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Células de Schwann/imunologia , Animais , Antígenos CD59/imunologia , Morte Celular/imunologia , Células Cultivadas , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/análise , Etídio/análogos & derivados , Etídio/análise , Humanos , Imunização Passiva/métodos , Substâncias Intercalantes/análise , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Microscopia Imunoeletrônica/métodos , Junção Neuromuscular/imunologia
17.
J Neuromuscul Dis ; 2(2): 157-165, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-27858734

RESUMO

BACKGROUND: Multifocal motor neuropathy (MMN) is associated with IgM antibodies to GM1 ganglioside. The importance of the lipid milieu that might facilitate or inhibit antibody binding to GM1 in immunoassays is well recognised. Existing studies, using a range of different approaches, generally concur that anti-GM1 IgM antibody detection rates are improved by the addition of galactocerebroside (GalC) to the GM1 assay. OBJECTIVE: The current study sought to formally evaluate the clinical utility of the GM1:GalC complex assay in the diagnosis of MMN. METHODS: Anti-GM1 and -GM1:GalC antibodies were examined using ELISA and glycoarray (dot blot) in a fully blinded study design, consisting of 100 MMN patients, 100 ALS cases and 100 healthy controls. RESULTS: The detection of anti-GM1 Abs using glycoarray was 67% sensitive and 85% specific. The addition of GalC to GM1, (1:1 weight to weight ratio), increased the sensitivity to 81% , whilst dropping specificity to 80% . Increasing the GalC content to a 1:5 ratio (or higher) further decreased specificity, and in doing so limited the usefulness of the GM1:GalC assay to the level of GM1 alone. The addition of GalC to the ELISA method also significantly increased sensitivity compared with GM1 alone, albeit with a significant decrease in specificity. CONCLUSIONS: This study indicates that the GM1:GalC assay is an advantageous assay adaptation for detecting anti-GM1 antibodies in MMN, using either glycoarray or ELISA, and warrants introduction into clinical diagnostic practice.

18.
Curr Opin Chem Biol ; 18: 78-86, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24495749

RESUMO

Humans with autoimmune peripheral neuropathies frequently harbour serum antibodies to single glycosphingolipids, especially gangliosides. Recently it has been appreciated that glycolipid and lipid complexes, formed from two or more individual species, can interact to create molecular shapes capable of being recognised by these autoantibodies whilst not binding to the single individuals. As a result of this, novel autoantibody targets have been identified. This newly termed 'combinatorial glycomic' approach has provided the impetus to redesigning the assay methodologies traditionally used in the neuropathy-associated autoantibody field. Combinatorial glycoarrays can be readily constructed in house using lipids of interest. Herein we especially highlight the role of the neutral lipids cholesterol and galactocerebroside in modifying glycosphingolipid orientation that subsequently favours or inhibits autoantibody binding.


Assuntos
Autoanticorpos/sangue , Doenças Autoimunes/imunologia , Glicômica/métodos , Glicoesfingolipídeos/imunologia , Doenças do Sistema Nervoso/imunologia , Animais , Epitopos/imunologia , Humanos
19.
Exp Neurol ; 233(2): 836-48, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22197826

RESUMO

Both the neural and glial components of the neuromuscular junction (NMJ) have been identified as potential sites for anti-ganglioside antibody (Ab) binding and complement-mediated injury in murine models for the human peripheral nerve disorder Guillain-Barré syndrome (GBS). Some patients suffering from the acute motor axonal neuropathy (AMAN) forms of GBS recover very rapidly from paralysis; it has been proposed that in these cases the injury was restricted to the distal motor axons and nerve terminals (NTs) which are able to regenerate over a very short time-frame. To test this hypothesis, the ventral neck muscles of mice (n=45) expressing cytosolic fluorescent proteins in their axons (CFP) and Schwann cells (GFP) were subjected to a single topical application of anti-ganglioside Ab followed by a source of complement. Group A (n=15) received Ab that selectively bound to the NTs, group B (n=15) received Abs that bound both to the NTs and the perisynaptic Schwann cells (pSCs) and group C (control animals; n=15) only received complement. Evolution of the injury was documented by in vivo imaging, and following euthanasia the muscles were reimaged ex vivo both quantitatively and qualitatively, either immediately, or after 1, 2, 3 or 5 days of regeneration (each n=3 per group). Within 15 minutes of complement application, a rapid loss of CFP overlying the NMJ could be seen; in group A, the GFP signal remained unchanged, whereas in group B the GFP signal was also lost. In group C no changes to either CFP or GFP were observed. At 24 h, 6% of the superficial NMJs in group A and 12% of the NMJs in group B exhibited CFP. In both groups, CFP returned within the next five days (group A: 93.5%, group B: 94%; p=0.739), with the recovery of CFP being preceded by a return of GFP-positive cells overlying the NMJ in group B. Auxiliary investigations revealed that the loss of CFP at the NMJ correlated with a loss of NT neurofilament immuno-reactivity and a return of CFP at the NMJ was accompanied by a return of neurofilament. In ultrastructural investigations, injured NTs were electron lucent and exhibited damaged mitochondria, a loss of filaments and a loss of synaptic vesicles. The examination of muscles after five days of regeneration revealed physiological NT-profiles. The results described above indicate that following a single anti-ganglioside Ab-mediated and complement-mediated attack, independent of whether there are healthy and mature perisynaptic Schwann cells overlying the NMJ, the murine NT is capable of recovering both its architectural and axolemmal integrity very rapidly. This data supports the notion that an equivalent mechanism may account for the rapid recovery seen in some clinical cases of AMAN.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Autoanticorpos/administração & dosagem , Proteínas do Sistema Complemento/toxicidade , Gangliosídeos/imunologia , Neurônios Motores/patologia , Junção Neuromuscular/lesões , Terminações Pré-Sinápticas/imunologia , Regeneração/fisiologia , Animais , Anticorpos Monoclonais/toxicidade , Autoanticorpos/toxicidade , Humanos , Camundongos , Camundongos Transgênicos , Neurônios Motores/imunologia , Músculos do Pescoço/imunologia , Músculos do Pescoço/patologia , Junção Neuromuscular/imunologia , Junção Neuromuscular/patologia , Regeneração/imunologia
20.
J Clin Invest ; 119(3): 595-610, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19221437

RESUMO

Anti-GM1 ganglioside autoantibodies are used as diagnostic markers for motor axonal peripheral neuropathies and are believed to be the primary mediators of such diseases. However, their ability to bind and exert pathogenic effects at neuronal membranes is highly inconsistent. Using human and mouse monoclonal anti-GM1 antibodies to probe the GM1-rich motor nerve terminal membrane in mice, we here show that the antigenic oligosaccharide of GM1 in the live plasma membrane is cryptic, hidden on surface domains that become buried for a proportion of anti-GM1 antibodies due to a masking effect of neighboring gangliosides. The cryptic GM1 binding domain was exposed by sialidase treatment that liberated sialic acid from masking gangliosides including GD1a or by disruption of the live membrane by freezing or fixation. This cryptic behavior was also recapitulated in solid-phase immunoassays. These data show that certain anti-GM1 antibodies exert potent complement activation-mediated neuropathogenic effects, including morphological damage at living terminal motor axons, leading to a block of synaptic transmission. This occurred only when GM1 was topologically available for antibody binding, but not when GM1 was cryptic. This revised understanding of the complexities in ganglioside membrane topology provides a mechanistic account for wide variations in the neuropathic potential of anti-GM1 antibodies.


Assuntos
Autoanticorpos/imunologia , Gangliosídeo G(M1)/imunologia , Glicolipídeos/fisiologia , Doenças do Sistema Nervoso Periférico/imunologia , Animais , Anticorpos Monoclonais/imunologia , Axônios/imunologia , Humanos , Camundongos , Neurônios Motores/imunologia , Terminações Nervosas/imunologia , Junção Neuromuscular/imunologia , Oligossacarídeos/imunologia , Transmissão Sináptica/imunologia , Proteína ran de Ligação ao GTP/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA