Next Article in Journal
Preventive Effects of Neuroprotective Agents in a Neonatal Rat of Photothrombotic Stroke Model
Next Article in Special Issue
Molecular Biomarkers in Multiple Sclerosis and Its Related Disorders: A Critical Review
Previous Article in Journal
A Phytochemical Constituent, (E)-Methyl-Cinnamate Isolated from Alpinia katsumadai Hayata Suppresses Cell Survival, Migration, and Differentiation in Pre-Osteoblasts
Previous Article in Special Issue
TNFAIP3 Deficiency Affects Monocytes, Monocytes-Derived Cells and Microglia in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neurological Syndromes Associated with Anti-GAD Antibodies

1
AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Neurologie 2-Mazarin, 75013 Paris, France
2
Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France
3
Neuroncology Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy
4
Department of Neuroscience, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(10), 3701; https://doi.org/10.3390/ijms21103701
Submission received: 27 April 2020 / Revised: 19 May 2020 / Accepted: 21 May 2020 / Published: 24 May 2020
(This article belongs to the Special Issue Molecular Biomarkers in Multiple Sclerosis and Its Related Disorders)

Abstract

:
Glutamic acid decarboxylase (GAD) is an intracellular enzyme whose physiologic function is the decarboxylation of glutamate to gamma-aminobutyric acid (GABA), the main inhibitory neurotransmitter within the central nervous system. GAD antibodies (Ab) have been associated with multiple neurological syndromes, including stiff-person syndrome, cerebellar ataxia, and limbic encephalitis, which are all considered to result from reduced GABAergic transmission. The pathogenic role of GAD Ab is still debated, and some evidence suggests that GAD autoimmunity might primarily be cell-mediated. Diagnosis relies on the detection of high titers of GAD Ab in serum and/or in the detection of GAD Ab in the cerebrospinal fluid. Due to the relative rarity of these syndromes, treatment schemes and predictors of response are poorly defined, highlighting the unmet need for multicentric prospective trials in this population. Here, we reviewed the main clinical characteristics of neurological syndromes associated with GAD Ab, focusing on pathophysiologic mechanisms.

1. Introduction

Glutamic acid decarboxylase (GAD) is an intracellular enzyme fairly expressed in neurons and insulin-secreting pancreatic β cells, whose physiologic function is the decarboxylation of glutamate to gamma-aminobutyric acid (GABA) [1,2].
GAD exists in two isoforms, GAD65 and GAD67, that share a similar structure consisting of an amino-terminal domain, a catalytic domain binding the cofactor pyridoxal 5’-phosphate (PLP), and a carboxy-terminal domain [3]. Despite a common structure, GAD65 and GAD67 differ with regard to several characteristics, including their amino acid sequence [3], their molecular weight [1], their localization within the cell, and their tonic enzymatic activity [4].
GAD67, encoded by the gene GAD1 on chromosome 2 (2q31.1) [5], is expressed early during embryogenesis [6] and has an essential role for the proper development of neural [7,8] and nonneural tissues [9]. In mature neurons, GAD67 is generally expressed in cell body and dendrites [10]. Being almost saturated with the PLP cofactor [4], GAD67 is constantly active and ensures the synthesis of basal levels of GABA [11].
GAD65, encoded by the gene GAD2 on chromosome 10 (10p12.1), is mainly expressed at the post-natal stage and is responsible for the rapid synthesis of GABA required for synaptic transmission [12]. GAD65 is primarily expressed in the pre-synaptic end of nerve terminals, where it exists in its inactive form, unbound to the PLP cofactor. By switching from the inactive to the active form [4,10], GAD65 allows a rapid and synthesis of GABA when needed.
Notwithstanding being an intracellular enzyme, pre-clinical studies have shown that GAD65 is able to associate with the plasma membrane [13] and surge to the extracellular space. Indeed, GAD65 is capable of anchor to the membrane of synaptic vesicles by forming a protein complex with other intracellular proteins, mechanism that ensures that GABA synthesis is coupled to its packaging in synaptic vesicles [13]. When synaptic vesicles fusion with the plasma membrane during exocytosis, GAD65 might consequently be transiently uncovered in the extracellular space [14]. The functional coupling between GABA and GAD65 are highlighted in Figure 1.

2. GAD Antibody Titers and Epitope Specificities

The autoantibodies commonly used in clinical practice recognize the GAD65 isoform of GAD. Although antibodies to the GAD67 isoform have been detected in the serum and the cerebrospinal fluid (CSF) of patients with various neurological syndromes [15,16,17], the latter are hardly ever detected in absence of GAD65 Ab [16,17,18] and thus are not considered clinically relevant. Differences in structure and surface electrostatic charges account for the lower autoantigenicity of GAD67 compared to GAD65 [11,19]. As most available evidence concerns GAD65 Ab, they will be thereafter simply indicated as GAD Ab.
Besides type 1 diabetes mellitus (T1DM) [20], GAD Ab have been associated with a number of neurological immune-mediated syndromes, including Stiff-Person Syndrome (SPS), cerebellar ataxia (CA), limbic encephalitis (LE) and temporal lobe epilepsy (TLE). This diversity of clinical manifestations reflects, at least in part, different epitope specificity: GAD Ab from diabetic patients seem to recognize distinctive epitopes in comparison to patients suffering with neurological syndromes, and GAD Ab from patients with SPS seem to recognize different epitopes than patients with CA or LE [21,22]. Nonetheless, there exists a massive overlap in epitope recognition, and not all studies have been able to highlight differences in epitope specificity [23]. Most of the epitopes of interest are located within the catalytic domain of the enzyme, although epitopes in the amino-terminal and carboxy-terminal domain have also been considered as potential Ab targets [15,21,22,23,24].
Patients with neurological syndromes have much higher titers of GAD Ab in serum than patients with T1DM [21,25,26], usually more than 100 fold greater, and they appear to stay high over time [27]. Some cut offs have been proposed for pathologic rates of GAD Ab: a high abnormal value can be defined by a Radioimmunoassay value greater than 2000 U/mL, by an ELISA value greater than 1000 IU/mL or 20 nmol/L, or by a strong positive labeling at low dilutions for immunohistochemistry. [12,28,29]. Ab titers might therefore be insufficient to distinguish whether the detection of GAD Ab in serum is in relation with the neurological syndrome or with an underlying T1DM. Nonetheless, only patients with GAD-related neurological syndromes will have GAD Ab detected in their CSF [28,30,31], and this represents a sturdy argument to establish an association between GAD Ab and neurological manifestations. In patients with neurological syndromes, no substantial differences were detected in GAD serum Ab titers between SPS, CA, and LE [15,21,26], supporting the concept that neurological phenotype is more likely dictated by epitope specificity rather than Ab titers. Furthermore, in previous studies, no correlation was found between the antibodies titers (serum and CSF) with the severity or duration [32] of the neurological disease. However, the anti-GAD Ab titers tend to decrease after immunotherapy [33,34,35].

3. GAD Ab Detection Strategies

In clinical practice, GAD Ab are detected through different techniques, including indirect immunohistochemistry [36,37], immunoblot [36], enzyme-linked immunosorbent assay (ELISA) [26,38], and radioimmunoassay (RIA) [28,36,38], which have different sensitivity and specificity.
Indirect immunohistochemistry is a method based on the incubation of patient sample on rat or primate brain sections. The incubation with a secondary antibody binding human IgG bound to a fluorophore or a colored marker allows then to detect the presence of autoantibodies in patient sample. The typical staining pattern associated with GAD Ab is characterized by a patchy staining of the granular layer of the cerebellum and a diffuse staining of the hippocampus [11,30,39,40,41]. A confirmatory test by line immunoblot will then allow to confirm the autoantibodies detected on immunohistochemistry as GAD Ab [38,42]. Immunohistochemistry is generally performed in laboratories dedicated to the detection of neuronal antibodies and, when used in combination with line assays, provides very accurate results [38].
ELISA is a technique widely used in laboratories of general immunology. GAD Ab are detected by using ELISA incubating patient sample in a well containing human recombinant GAD65. A secondary antibody, linked to an enzyme, is then added to the well: if the patient sample harbors GAD Ab, a change of color will arise as a consequence of enzymatic activity [43]. The chromatic change is then quantified by computerized plate readers [15].
RIA is a technique that allows the detection of GAD Ab by precipitating patient sample with human recombinant GAD65 labelled with Iodine-125 (125I) [38,44]. The precipitate is then counted for 125I using a gamma counter. Although not broadly used, this technique represents the standard for GAD Ab detection in some expertise laboratories [45].
Although more sensitive than indirect immunohistochemistry and line assays, ELISA and RIA can be associated with false positive results. Clinicians should therefore be extremely cautious in the interpretation of low Ab titers detected using these techniques [38].

4. Physiopathology: Decreased GABAergic Transmission

GABAergic neurons are part of a large network of inhibitory interneurons responsible for inhibitory signals throughout the central nervous system, primarily located in the hippocampus, the cerebellum, basal ganglia, brainstem nuclei, and spinal gray matter [46,47]. GABAergic neurons express high levels of GAD65, the enzyme responsible for GABA synthesis. After its synthesis in pre-synaptic terminals, GABA is packed within synaptic vesicles and liberated in the synaptic cleft through a mechanism of exocytosis [13]. Released in the synaptic cleft, GABA binds the GABAA and GABAB receptors, whose activation results in a hyperpolarization of post-synaptic neurons and thus in an inhibitory signal [48,49].
Although the underlying mechanisms have not been fully elucidated yet, it has been extensively demonstrated that GAD autoimmunity interferes with GABAergic synaptic transmission. GAD Ab have been hypothesized to have an inhibitory effect on GAD65, blocking GABA synthesis by reducing the uptake of newly-synthetized GABA in synaptic vesicles and its synaptic release [13,50,51,52]. In GAD65 knockout mice, GABAergic release is impaired only after repetitive stimulations [53]. The decreased synthesis of GABA would in turn result in decreased GABAergic transmission and thus in a state of neuronal hyperexcitability [54,55] similar to the one observed in pre-clinical models of functional or genetic inactivation of GABA receptors [9,56,57]. Altered GABAergic transmission is indeed the core pathophysiological mechanism in SPS: the reduced firing of inhibitory GABAergic neurons in the spinal cord causes a state of nerve hyperexcitability of motor neurons, leading to the simultaneous contraction of agonist and antagonist muscles that is typical of SPS [11,58]. Further evidence suggests an additional involvement of GABAergic interneurons at the supraspinal level, with an hyperexcitability of cortical motor neurons [52]. A recent study has shown that patients with SPS have lower levels of GABA in their CSF compared to control subjects [30], providing a biological correlate to neurophysiological observations.
Functional alterations in GABAergic transmission are also supposed to underlie the pathogenesis of GAD-associated CA [59]. GAD Ab from patients with CA incubated on cerebellar slices in vitro have shown to decrease the pre-synaptic release of GABA, and extensively in Purkinje cells [54,60,61], this effect being partially reversible upon the use of Forskolin, an activator of cAMP [62]. In vivo studies in rodents have shown that the intracerebellar administration of GAD IgG from affected patients blocked the potentiation of corticomotor response [63]. This evidence supports the hypothesis that, similar to SPS, GAD autoimmunity results in a reduced synthesis and release of GABA from pre-synaptic neurons and thus in a decrease of inhibitory signals downstream [54,55,59,60,61,64,65].
Limited data are available on the alterations of GABAergic transmission in GAD-associated LE and temporal lobe epilepsy. In vitro studies have shown that GAD Ab from patients with epilepsy result in a significant increase in post-synaptic inhibitory potentials in cultured hippocampal neurons, suggesting an interference with GABA function [66]. Furthermore, in vivo evidence supports the evidence that reduced GABAergic transmission results in cortical hyperexcitability, GAD65 knockout mice developing seizures involving limbic regions [9,52].

5. Immune Effectors and Pathogenetic Mechanisms

While it is clear that altered GABAergic transmission represents the core of physiopathology, it remains debated whether the effectors of GAD autoimmunity are GAD Ab per se or the accompanying T cell response. In type 1 diabetes associated with GAD autoimmunity, CD4+ and CD8+ T cell responses both seem involved [67,68,69].
Antibodies to neural intracellular antigens, such as onconeural antibodies, are generally considered surrogate markers of T cell activation lacking a direct pathogenic role [70]. Certainly, some evidence shows that GAD autoimmunity might mostly be cell-mediated. Monoclonal GAD65-specific CD4+ T cell populations have proven able to cause lethal encephalomyelitis in mice in absence of other effectors [71]. Inflammatory infiltrates of CD8+ T lymphocytes were detected in the spinal cord of patients deceased of SPS, together with axonal swelling and neuronal loss [72,73,74]. Similar findings were detected in a post-mortem study of a patient deceased because of CA, the selective loss of Purkinje cells pointing to a cell-mediated cytotoxic response [75]. It is assumed that the inflammatory cascade induced by GAD Ab is responsible of lesions leading to neuronal loss and cerebellar atrophy in chronic impairment. Evidence of T cell autoimmunity is also available for GAD-related LE, with post-mortem studies showing cytotoxic T cells in close proximity to neurons and severe neuronal loss in the hippocampus [76].
Nonetheless, the ability of GAD to surface to the extracellular space during the exocytosis of synaptic vesicles has prompted some authors to postulate that GAD Ab might be directly pathogenic. Differently from other antibodies to intracellular antigens, GAD Ab might in fact directly interact with their target antigen [15]. The intrathecal injection of GAD Ab from patients with SPS or CA proved sufficient to reproduce typical neurophysiological manifestations in animal models [59,63], supporting the hypothesis that GAD Ab might have a pathogenic role.
This conflicting evidence hampers to draw precise conclusions on the immune mechanisms responsible for neuronal dysfunction in GAD autoimmunity [12,14], highlighting to what extent GAD Ab constitute a peculiar case among antibodies to intracellular antigens.

6. Epidemiology

GAD-related neurological syndromes are uncommon, the estimated prevalence being 1/1,250,000 for SPS [77] and 1.9/100,000 for LE [78].
GAD Ab are detected in about 60%–80% of cases of classical SPS [45,79], the remaining proportion of SPS cases being seronegative or associated with other neural Ab (amphiphysin, glycine receptor) [45,58]. GAD Ab account for about 2% of sporadic progressive CA [80] and for 12% of CA of unknown origin [81], making it a non-negligible cause of cerebellar dysfunction. They are also present in the serum of approximately 17% of patients with LE [29] and in 2.1%–5.4% of patients with unexplained epilepsy (considering only high Ab titers) [29,82,83,84,85].
Certainly, GAD Ab can be detected in the serum of 0.4%–1.7% of healthy subjects [16,84,86,87], in the serum of 5% of patients with neurological disorders of other etiology [16], and in the serum of 80% of newly diagnosed patients with T1DM [25], highlighting the importance to interpret the results of serum testing in the light of neurological presentation and alternative etiologies, demanding CSF testing in ambiguous cases.
Neurological syndromes related to GAD autoimmunity display a sharp female prevalence, with a percentage of women > 80% in all main phenotypes [15,28]. The median age at disease onset is between 50 and 60 years old for SPS and CA [15,28,45,87], while is between 25 and 45 years for LE [15,29,88].

7. Genetic Predisposition

Similar to other autoimmune disorders, GAD autoimmunity has been associated with both genetic and environmental risk factors. The genes encoding for the human leukocyte antigen (HLA) system, on chromosome 6, have been the ones most studied thus far, due to their recognized relation with autoimmunity. Curiously, despite all being related to GAD Ab, T1DM and neurological syndromes seem associated with different HLA haplotypes, strengthening the hypothesis that these two manifestations are triggered and supported by different immunological mechanisms. GAD Ab positive T1DM has been associated with the HLA class II haplotypes DQA1*05:01-DQB1*02:01- DRB1*03:01 (DQ2-DR3) and DQA1*03:01-DQB1*03:02- DRB1*04 (DQ3-DR4) [89,90], the DQ2-DR3 haplotype being also related to autoimmune thyroiditis [91]. Neurological syndromes have instead been associated the HLA class II haplotype DQA1*05:01–DQB1*02:01–DRB1*03:01 (DQ2–DR3), detected in 41% of patients in a mixed cohort of SPS, CA, and LE [89]. Consistently, the HLA alleles DQB1*02:01 and DRB1*03:0 were formerly detected in 44%–72% of patients in cohorts with SPS [92,93]. Further studies analyzing larger cohorts are needed to clarify whether the different neurological syndromes are associated with particular HLA alleles.

8. Coexisting Autoimmune Disorders

Patients with neurological disorders associated with GAD Ab typically have a personal or familial history of autoimmunity. Family history of autoimmune diseases, including T1DM or thyroiditis, is common (43%–55% for SPS/CA [79,87] and 23% for TLE [93]). A personal history of T1DM is present in about half of cases (between 43 and 71% in the main cohorts [15,18,28,45,87]) and it usually precedes neurological manifestations [28,45,87]. Besides T1DM, the majority of patients will present one or more additional manifestations of systemic autoimmunity, including thyroiditis, pernicious anemia, coeliac disease, and vitiligo, reflecting the presence of anti-thyroid peroxidase, anti-thyroglobulin, anti-parietal cells, and anti-gliadin antibodies [15,28,29,45,87,88]. These antecedents seem more common in patients with CA and LE (50%–70%) than in patients with SPS (30%–50%) [15,28,29,45,87,88]. These data emphasize to what extent GAD autoimmunity is associated with other systemic manifestations of autoimmunity [11], encouraging further research on common predisposing factors.

9. Association with Cancer

Paraneoplastic cases constitute a minor percentage of the neurological syndromes associated with GAD Ab. Only 4%–6% of patients with GAD positive SPS have associated malignant tumors [45,77,94] (mostly thymomas [94,95], and breast [96,97], thyroid, renal, and colon cancer [45]). The association with cancer seems relatively more frequent in CA and LE, with a reported percentage of paraneoplastic cases of 9% and 26%, respectively [98]. Underlying cancer types also differ compared to SPS, non-small cell lung cancer and pancreatic neuroendocrine tumors being the most common [28].
Older age, male gender, and coexisting Ab to cell surface antigens (e.g., GABA or glycine receptor Ab) [98,99] represent recognized risk factors for an underlying malignancy and should prompt regular tumor screening, which is not generally encouraged in patients with GAD-related neurological syndromes due to the low association with cancer.
Although cancer association is certainly unusual in GAD autoimmunity, several studies have demonstrated that this event is not coincidental but reflects a paraneoplastic mechanism. Immunohistochemical studies conducted on neuroendocrine tumors from patients diagnosed with GAD-associated CA have proven that tumor cells display a strong GAD expression [100,101] responsible for the immune cross-reaction with cerebellar neurons.
Interestingly, GAD autoimmunity is almost never reported in association with paraneoplastic neuronal antibodies [40].

10. Other Triggers of Autoimmunity

The use of immune checkpoint inhibitors (ICI) for cancer immunotherapy has been associated with a variety of immune-related neurological adverse events, including meningoencephalitis, polyradiculoneuritis, myositis, and myasthenia gravis [102]. Neurological complications are uncommon, estimated to occur in about 1% of patients treated with ICI, and only rarely associated with autoantibodies [102]. GAD Ab have been detected in few patients developing LE following the administration of ICIs (nivolumab and ipilimumab) for lung or thymic carcinoma [103,104], although, unfortunately, GAD expression was not researched in tumor specimen to circumstantiate the paraneoplastic nature of autoimmunity. On the other hand, some cases of diabetic ketoacidosis due to a fulminant autoimmune T1DM associated with GAD Ab have been reported in patients with no neurological symptoms or concomitant autoimmune diseases [105,106,107].

11. Neurological Presentation

The main clinical and paraclinical features associated with GAD-related neurological syndromes are reported hereafter and summarized in Table 1.

11.1. Stiff-Person Syndrome

SPS was first described in 1956 in a small series of 14 cases published by Moersch and Woltman [108]. The classical phenotype associated with SPS includes axial rigidity predominating in the trunk and in proximal lower limbs, accompanied by painful muscle spasms that might cause falls or respiratory difficulties due to their interference with normal muscle contraction [45,79]. Muscle spasms may be induced by unexpected tactile stimuli, sudden noises, and emotional stress and are often accompanied by symptoms such as anxiety, obsession with details and phobias [79]. On neurological examination, lumbar hyperlordosis and co-contracture of paraspinal and abdominal muscles is a common and distinguishing characteristic [79,109]. In about one third of patients, SPS has a focal presentation, with rigidity and painful spasms limited to the trunk or to one limb (i.e., “focal” SPS) [45]. GAD antibodies are detected in 88%–98% of patients with classical SPS [44,45,108] and in 15%–61% of patients with “focal” SPS [45,108].
The diagnosis of SPS is often hard to attain and patients commonly have a long-standing history of unfruitful investigations [79], leading to suspect a functional disorder. Neurophysiological examinations are extremely important to circumstantiate patients’ complaints. In about 60% of patients with SPS, signs of spinal hyperexcitability are detected on the electromyogram (EMG) [45], including continuous motor unit activity, exaggerated exteroceptive reflexes, and abnormal cutaneomuscular reflexes [77,109,110]. Although this percentage might appear quite low, the continued use of benzodiazepines or baclofen might explain the negativity of the EMG in a substantial proportion of cases [45]. Brain and spine magnetic resonance imaging (MRI) are usually normal [109] and are generally performed to exclude alternative causes. CSF analysis commonly shows normal protein levels and cell count [109]. CSF-specific oligoclonal bands are frequently detected [15,28,109] and seem more common in patients with classical (57%–66%) [30,109] instead of focal (17%) SPS [109]. An intrathecal synthesis of GAD Ab is reported in 82%–85% of cases [28,79]. The detection of GAD Ab in the CSF, even more so if supported by the presence of oligoclonal bands and/or intrathecal synthesis of Ab, represents a robust argument in favor of a diagnosis of SPS, especially in atypical cases [77].

11.2. Cerebellar Ataxia

CA is another common syndrome associated with GAD autoimmunity. The most common symptoms include gait ataxia, dysarthria, and nystagmus [87], corresponding to a primary involvement of cerebellar vermis. Symptom onset is commonly subacute or chronic, with an insidious progression over months or years [18,28,87]. It is estimated that, at the nadir of disease severity, 71% patients have a modified Rankin Score (mRS) ≥ 3, corresponding to the need of using unilateral support for walking or to a wheelchair-bound condition [87].
Brain MRI reveals an atrophy of the cerebellum, and especially of the vermis, in 43% to 57% of patients [18,81,111,112]. Similar to other immune-mediated CA, cerebellar atrophy is not present at disease onset but usually develops over time, becoming frequent and, possibly, severe in patients with a long-standing symptom evolution [81,87,112]. Positron emission tomography (PET) studies performed in patients with cerebellar atrophy show a correspondent cerebellar hypometabolism [113] confirming neuronal degeneration.
CSF analysis commonly shows normal protein levels and cell count but evidence of intrathecal synthesis of GAD Ab (83%–100%) and CSF-specific oligoclonal bands (69%–73%) [18,28,87].

11.3. Limbic Encephalitis and Autoimmune Epilepsy

GAD Ab have been consistently associated with LE and TLE [12,85], which probably represent overlapping entities [93]. Similar to other types of LE, GAD-associated LE is characterized by the subacute onset of seizures (53%) [93], anterograde amnesia (67%), and confusion and behavioral changes (30%) [93,114]. While virtually all patients will experience seizures at some point [93], status epilepticus is rarely reported [115,116]. Almost all patients experience cognitive impairment on the long term, with an impairment of memory and frontal lobe function [93]. Hyponatremia, another distinguishing hallmark of limbic encephalitis, is rarely reported.
Brain MRI typically pinpoints bilateral hyperintensities in temporo-mesial structures on T2 weighted or fluid-attenuated inversion recovery (FLAIR) sequences, with no contrast enhancement [93]. Although not usually present at diagnosis, medio-temporal cortical atrophy usually becomes evident within 6 to 12 months [12,76]. PET studies commonly show an hypermetabolism of mesiotemporal cortex during the early (inflammatory) phase, while a correspondent hypometabolism, reflecting neuronal loss or dysfunction, generally becomes apparent at the chronic (atrophic) stage [29,84].
Electroencephalograms (EEG) frequently show slow or epileptic anomalies over the temporal regions, ictal and interictal discharges being evident in two-thirds of patients with GAD-associated epilepsy [12].
CSF analysis can display inflammatory findings, with a moderate pleocytosis (1–25 cells/L) (6–36%) [29,93] and a mild elevation in protein levels (0.4–0.9 g/L) [29,76]. CSF-specific oligoclonal bands (63%–100%) and intrathecal synthesis of GAD Ab are common [15,28,29,93].
Besides LE, GAD Ab have been detected in the serum and CSF of patients with long-standing or refractory temporal lobe [12,93] or generalized [84] epilepsy. While indeed some patients had evidence of temporal lobe involvement with GAD Ab and/or oligoclonal bands detected in their CSF, supporting an autoimmune origin of their epilepsy, others did not have these features [84,85], rending questionable the autoimmune origin of their epilepsy [88]. The detection of GAD Ab in the CSF and/or the presence of an intrathecal synthesis of IgG represent capital elements to support a causal relation between GAD Ab and neurological manifestations [12], and should always be carefully researched.

11.4. Overlap Syndromes

Overlap syndromes are commonly observed during follow-up and, over the years, patients can develop combinations of SPS, CA, or LE. Overlap syndromes are observed in 10%–20% of patients with SPS [15,45,117], in 14%–36% of patients with CA [15,18,28,87], and in 10%–25% of patients with LE/epilepsy [15,29,88]. Less commonly, patients display signs of involvement of other neurological structures or systems that are not usually affected by GAD autoimmunity, such as the brainstem and basal ganglia, causing oculomotor disorders or extrapyramidal symptoms [40].

11.5. Other Neurological Syndromes Associated with GAD Ab

Besides the three main syndromes, GAD Ab have been associated with other neurological manifestations, including progressive encephalomyelitis with rigidity and myoclonus (PERM) [45,118,119], opsoclonus-myoclonus [120], palatal tremor [121,122], myelitis [123], and autonomic neuropathy [124]. GAD Ab have also been reported in the serum of patients affected by juvenile neuronal ceroid lipofuscinosis (Batten disease), a genetic neurodegenerative disease manifesting with altered psychomotor development, impaired vision, and seizures. These patients also had evidence of GABAergic neuron loss at post-mortem studies [125].

12. Treatment and Outcome

Similar to other autoimmune conditions, the treatment of GAD-associated neurological syndromes has not been codified yet, and the schemes of immune therapy are highly dependent on the experience of individual centers and physicians. Treatment and outcome in the main series reported in literature for each of the three main neurological syndromes are reported in Table 2.

12.1. Stiff-Person Syndrome

In patients with SPS, symptomatic treatment often represents the first line of therapy, as it allows to achieve a resolution or, at least, an improvement of symptoms in the majority of patients (78%–100%) [45,109]. Benzodiazepines (e.g., diazepam, clonazepam) and baclofen (oral or intrathecal) generally represent the first choice due to their recognized GABAergic action, although other drugs, including antiepileptics (e.g., valproate, gabapentin, levetiracetam), drugs with anti-monoaminergic effects (clonidine and tizanidine), dantrolene and injections of botulinum toxin, have also been occasionally reported as effective [45,77].
Patients not responding adequately to symptomatic treatment are usually proposed an immune therapy based on intravenous immunoglobulin (IVIg), plasma exchange (PE), or high-dose corticosteroids. A randomized controlled trial on 16 patients with SPS showed a significant improvement of stiffness and spasms in patients receiving IVIg compared to placebo, accompanied by a substantial functional improvement and a decrease of GAD Ab titers in serum [33]. PE [45,127,138] and high-dose corticosteroids [45] have been used in individual cases, allowing to achieve some improvement or, at least, stability.
Rituximab [34,128,139] and intravenous cyclophosphamide [45] are used in patients who are severely disabled and show no response to first-line treatments. In case maintenance treatment is needed, monthly courses of IVIg or PE, a protracted course of oral corticosteroids or immune suppressants such as azathioprine or mycophenolate mofetil can be used for prolonged immune modulation [45]. The reported benefits of immunotherapy differ among studies, with percentages of improvement ranging from 39% to 80% of cases [26,45]. Despite some improvement, a substantial proportion of patients are left moderately disabled, 30%–57% requiring support for walking or being wheelchair-bound [26,45].

12.2. Cerebellar Ataxia

Patients with CA invariably need one or more lines of immunotherapy, as the absence of treatment usually results in further clinical deterioration [18]. First-line immunotherapy is based on the administration of IVIg, high-dose intravenous corticosteroids, PE, or rituximab, used alone or in combination [140]. Most patients will then need a maintenance therapy consisting of monthly courses of IVIg, oral corticosteroids, azathioprine, or mycophenolate mofetil [18].
As no clinical trials have been conducted in this population, data on treatment response and outcome are limited to retrospective series. A large study on 34 patients with CA, showed that immunotherapy resulted in an improvement, in terms of modified Rankin Score (mRS), in about 50% of patients [18]. Patients with subacute onset and patients with shorter delay to immunotherapy administration had better response to treatment [18], highlighting the importance of early diagnosis and immune therapy administration. Despite some improvement after immunotherapy, half of the patients experience moderate to severe long-term sequelae, with a mRS ≥ 3 at last follow-up, corresponding to the need of unilateral support for walking or a wheelchair-bound condition [18,140].

12.3. Limbic Encephalitis and Autoimmune Epilepsy

The first-line treatment of LE relies on the administration of IVIg, high-dose intravenous corticosteroids, or PE, alone or in combination [12], followed by a rapid switch to rituximab or cyclophosphamide in absence of satisfactory clinical response. Improvement rates after immunotherapy administration are usually modest [12,26,29,93,137] and most patients continue experiencing seizures (80%) [93] and cognitive impairment (69%) [93]. In a cohort of patients with LE, only 18% of patients with GAD Ab were seizure free after receiving immunotherapy compared to 55% of patients with paraneoplastic LE and other antibodies [141], suggesting that GAD-related epilepsy is particularly difficult to control. Seizures are often resistant to antiepileptic drugs and, despite limited expectations, immunotherapy still provides the best chances to control epileptic manifestations [12,26].
Similar observations are true for TLE associated with GAD Ab [137]. A recent study on 35 patients with temporal lobe epilepsy and GAD Ab in the CSF showed that, despite immunotherapy, most of the patients developed refractory epilepsy and progressive cognitive impairment [93], highlighting the striking treatment-resistance of this condition.
In paraneoplastic cases, oncological treatment is essential, in parallel to immunotherapy, to improve neurological outcome.

Author Contributions

Conceptualization, M.D., G.B., and A.A.; data curation, M.D. and G.B.; writing—original draft preparation, M.D., G.B., and A.A.; writing—review and editing, M.D., G.B., C.I., M.G., M.B., J.-Y.D., D.P., and A.A.; visualization, A.A.; supervision, G.B. and A.A.; project administration, M.D. and A.A.; funding acquisition, A.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been developed within the BETPSY project, which is supported by a public grant overseen by the French National Research Agency (ANR) as part of the second “Investissements d’Avenir” program (reference ANR-18-RHUS-0012).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Abbreviations

GADGlutamic acid decarboxylase
GABAGamma-aminobutyric acid
PLPPyridoxal 5’-phosphate
AbAntibodies
SvsSynaptic vesicles
HCS70Heat Shock Cognate 70
CSPCysteine-String Protein
VGATVesicular GABA transporter
CaMKIICalcium/calmoduline protein kinase
T1DMType 1 diabetes mellitus
SPSStiff-person syndrome
CACerebellar ataxia
LELimbic encephalitis
CSFCerebrospinal fluid
ELISAEnzyme-linked immunosorbent assay
RIARadioimmunoassay
125IIodine-125
SVSynaptic vesicles
HLAHuman leukocyte antigen
TLETemporal lobe epilepsy
ICIImmune checkpoint inhibitors
EMGElectromyogram
MRImagnetic resonance imaging
MDSMyelodysplastic syndrome
PETPositron emission tomography
PAPernicious anemia
NSCLCNon-small cell lung cancer
NANot available
PERMProgressive encephalomyelitis with rigidity and myoclonus
mRSModified Rankin Score
FLAIRFluid-attenuated inversion recovery
EEGElectroencephalogram
CSCorticosteroids
FUFollow-up
IVIntravenous
IVIgIntravenous immunoglobulin
MPMethylprednisolone
PEPlasma exchange
ICARSInternational Cooperative Ataxia Rating Scale
AZAAzathioprine

References

  1. Solimena, M.; De Camilli, P. Autoimmunity to glutamic acid decarboxylase (GAD) in Stiff-Man syndrome and insulin-dependent diabetes mellitus. Trends Neurosci. 1991, 14, 452–457. [Google Scholar] [CrossRef]
  2. Vincent, S.R.; Hökfelt, T.; Wu, J.Y.; Elde, R.P.; Morgan, L.M.; Kimmel, J.R. Immunohistochemical studies of the GABA system in the pancreas. Neuroendocrinology 1983, 36, 197–204. [Google Scholar] [CrossRef] [PubMed]
  3. Martin, D.L.; Rimvall, K. Regulation of Gamma-Aminobutyric Acid Synthesis in the Brain. Available online: https://pubmed-ncbi-nlm-nih-gov.proxy.insermbiblio.inist.fr/8419527/?from_term=martin+neurochem+1993&from_pos=1 (accessed on 26 February 2020).
  4. Kaufman, D.L.; Houser, C.R.; Tobin, A.J. Two forms of the gamma-aminobutyric acid synthetic enzyme glutamate decarboxylase have distinct intraneuronal distributions and cofactor interactions. J. Neurochem. 1991, 56, 720–723. [Google Scholar] [CrossRef]
  5. Bu, D.F.; Tobin, A.J. The exon-intron organization of the genes (GAD1 and GAD2) encoding two human glutamate decarboxylases (GAD67 and GAD65) suggests that they derive from a common ancestral GAD. Genomics 1994, 21, 222–228. [Google Scholar] [CrossRef] [PubMed]
  6. Popp, A.; Urbach, A.; Witte, O.W.; Frahm, C. Adult and embryonic GAD transcripts are spatiotemporally regulated during postnatal development in the rat brain. PLoS ONE 2009, 4, e4371. [Google Scholar] [CrossRef] [PubMed]
  7. Chattopadhyaya, B.; Di Cristo, G.; Wu, C.Z.; Knott, G.; Kuhlman, S.; Fu, Y.; Palmiter, R.D.; Huang, Z.J. GAD67-mediated GABA Synthesis and Signaling Regulate Inhibitory Synaptic Innervation in the Visual Cortex. Neuron 2007, 54, 889–903. [Google Scholar] [CrossRef] [Green Version]
  8. Magri, C.; Giacopuzzi, E.; La Via, L.; Bonini, D.; Ravasio, V.; Elhussiny, M.E.A.; Orizio, F.; Gangemi, F.; Valsecchi, P.; Bresciani, R.; et al. A novel homozygous mutation in GAD1 gene described in a schizophrenic patient impairs activity and dimerization of GAD67 enzyme. Sci Rep. 2018, 8. [Google Scholar] [CrossRef] [Green Version]
  9. Kash, S.F.; Johnson, R.S.; Tecott, L.H.; Noebels, J.L.; Mayfield, R.D.; Hanahan, D.; Baekkeskov, S. Epilepsy in mice deficient in the 65-kDa isoform of glutamic acid decarboxylase. Proc. Natl. Acad. Sci. USA 1997, 94, 14060–14065. [Google Scholar] [CrossRef] [Green Version]
  10. Erlander, M.G.; Tillakaratne, N.J.; Feldblum, S.; Patel, N.; Tobin, A.J. Two genes encode distinct glutamate decarboxylases. Neuron 1991, 7, 91–100. [Google Scholar] [CrossRef]
  11. McKeon, A.; Tracy, J.A. GAD65 neurological autoimmunity. Muscle Nerve 2017, 56, 15–27. [Google Scholar] [CrossRef]
  12. Daif, A.; Lukas, R.V.; Issa, N.P.; Javed, A.; VanHaerents, S.; Reder, A.T.; Tao, J.X.; Warnke, P.; Rose, S.; Towle, V.L.; et al. Antiglutamic acid decarboxylase 65 (GAD65) antibody-associated epilepsy. Epilepsy Behav. 2018, 80, 331–336. [Google Scholar] [CrossRef] [PubMed]
  13. Jin, H.; Wu, H.; Osterhaus, G.; Wei, J.; Davis, K.; Sha, D.; Floor, E.; Hsu, C.-C.; Kopke, R.D.; Wu, J.-Y. Demonstration of functional coupling between gamma -aminobutyric acid (GABA) synthesis and vesicular GABA transport into synaptic vesicles. Proc. Natl. Acad. Sci. USA 2003, 100, 4293–4298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lancaster, E.; Dalmau, J. Neuronal autoantigens—Pathogenesis, associated disorders and antibody testing. Nat. Rev. Neurol. 2012, 8, 380–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Gresa-Arribas, N.; Ariño, H.; Martínez-Hernández, E.; Petit-Pedrol, M.; Sabater, L.; Saiz, A.; Dalmau, J.; Graus, F. Antibodies to inhibitory synaptic proteins in neurological syndromes associated with glutamic acid decarboxylase autoimmunity. PLoS ONE 2015, 10, e0121364. [Google Scholar] [CrossRef] [PubMed]
  16. Meinck, H.M.; Faber, L.; Morgenthaler, N.; Seissler, J.; Maile, S.; Butler, M.; Solimena, M.; DeCamilli, P.; Scherbaum, W.A. Antibodies against glutamic acid decarboxylase: Prevalence in neurological diseases. J. Neurol. Neurosurg. Psychiatry 2001, 71, 100–103. [Google Scholar] [CrossRef] [Green Version]
  17. Guasp, M.; Solà-Valls, N.; Martínez-Hernández, E.; Gil, M.P.; González, C.; Brieva, L.; Saiz, A.; Dalmau, J.; Graus, F.; Ariño, H. Cerebellar ataxia and autoantibodies restricted to glutamic acid decarboxylase 67 (GAD67). J. Neuroimmunol. 2016, 300, 15–17. [Google Scholar] [CrossRef] [Green Version]
  18. Ariño, H.; Gresa-Arribas, N.; Blanco, Y.; Martínez-Hernández, E.; Sabater, L.; Petit-Pedrol, M.; Rouco, I.; Bataller, L.; Dalmau, J.O.; Saiz, A.; et al. Cerebellar Ataxia and Glutamic Acid Decarboxylase Antibodies: Immunologic Profile and Long-term Effect of Immunotherapy. JAMA Neurol. 2014, 71, 1009. [Google Scholar] [CrossRef] [Green Version]
  19. Fenalti, G.; Buckle, A.M. Structural biology of the GAD autoantigen. Autoimmun. Rev. 2010, 9, 148–152. [Google Scholar] [CrossRef]
  20. Reetz, A.; Solimena, M.; Matteoli, M.; Folli, F.; Takei, K.; De Camilli, P. GABA and pancreatic beta-cells: Colocalization of glutamic acid decarboxylase (GAD) and GABA with synaptic-like microvesicles suggests their role in GABA storage and secretion. EMBO J. 1991, 10, 1275–1284. [Google Scholar] [CrossRef] [Green Version]
  21. Manto, M.; Honnorat, J.; Hampe, C.S.; Guerra-Narbona, R.; López-Ramos, J.C.; Delgado-García, J.M.; Saitow, F.; Suzuki, H.; Yanagawa, Y.; Mizusawa, H.; et al. Disease-specific monoclonal antibodies targeting glutamate decarboxylase impair GABAergic neurotransmission and affect motor learning and behavioral functions. Front. Behav. Neurosci. 2015, 9, 78. [Google Scholar] [CrossRef] [Green Version]
  22. Raju, R.; Foote, J.; Banga, J.P.; Hall, T.R.; Padoa, C.J.; Dalakas, M.C.; Ortqvist, E.; Hampe, C.S. Analysis of GAD65 autoantibodies in Stiff-Person syndrome patients. J. Immunol. 2005, 175, 7755–7762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Fouka, P.; Alexopoulos, H.; Akrivou, S.; Trohatou, O.; Politis, P.K.; Dalakas, M.C. GAD65 epitope mapping and search for novel autoantibodies in GAD-associated neurological disorders. J. Neuroimmunol. 2015, 281, 73–77. [Google Scholar] [CrossRef] [PubMed]
  24. Burbelo, P.D.; Groot, S.; Dalakas, M.C.; Iadarola, M.J. High Definition Profiling of Autoantibodies to Glutamic Acid Decarboxylases GAD65/GAD67 in Stiff-Person Syndrome. Biochem. Biophys. Res. Commun. 2008, 366, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Baekkeskov, S.; Aanstoot, H.-J.; Christgai, S.; Reetz, A.; Solimena, M.; Cascalho, M.; Folli, F.; Richter-Olesen, H.; Camilli, P.-D. Identification of the 64K autoantigen in insulin-dependent diabetes as the GABA-synthesizing enzyme glutamic acid decarboxylase. Nature 1990, 347, 151–156. [Google Scholar] [CrossRef]
  26. Muñoz-Lopetegi, A.; de Bruijn, M.A.A.M.; Boukhrissi, S.; Bastiaansen, A.E.M.; Nagtzaam, M.M.P.; Hulsenboom, E.S.P.; Boon, A.J.W.; Neuteboom, R.F.; de Vries, J.M.; Sillevis Smitt, P.A.E.; et al. Neurologic syndromes related to anti-GAD65: Clinical and serologic response to treatment. Neurol. Neuroimmunol. Neuroinflamm. 2020, 7. [Google Scholar] [CrossRef] [Green Version]
  27. Nakajima, H.; Nakamura, Y.; Inaba, Y.; Tsutsumi, C.; Unoda, K.; Hosokawa, T.; Kimura, F.; Hanafusa, T.; Date, M.; Kitaoka, H. Neurologic disorders associated with anti-glutamic acid decarboxylase antibodies: A comparison of anti-GAD antibody titers and time-dependent changes between neurologic disease and type I diabetes mellitus. J. Neuroimmunol. 2018, 317, 84–89. [Google Scholar] [CrossRef]
  28. Saiz, A.; Blanco, Y.; Sabater, L.; González, F.; Bataller, L.; Casamitjana, R.; Ramió-Torrentà, L.; Graus, F. Spectrum of neurological syndromes associated with glutamic acid decarboxylase antibodies: Diagnostic clues for this association. Brain 2008, 131, 2553–2563. [Google Scholar] [CrossRef] [Green Version]
  29. Malter, M.P.; Helmstaedter, C.; Urbach, H.; Vincent, A.; Bien, C.G. Antibodies to glutamic acid decarboxylase define a form of limbic encephalitis. Ann. Neurol. 2010, 67, 470–478. [Google Scholar] [CrossRef]
  30. Dalakas, M.C.; Li, M.; Fujii, M.; Jacobowitz, D.M. Stiff person syndrome: Quantification, specificity, and intrathecal synthesis of GAD65 antibodies. Neurology 2001, 57, 780–784. [Google Scholar] [CrossRef]
  31. Jarius, S.; Stich, O.; Speck, J.; Rasiah, C.; Wildemann, B.; Meinck, H.M.; Rauer, S. Qualitative and quantitative evidence of anti-glutamic acid decarboxylase-specific intrathecal antibody synthesis in patients with stiff person syndrome. J. Neuroimmunol. 2010, 229, 219–224. [Google Scholar] [CrossRef]
  32. Rakocevic, G.; Raju, R.; Dalakas, M.C. Anti-glutamic acid decarboxylase antibodies in the serum and cerebrospinal fluid of patients with Stiff-Person syndrome: Correlation with clinical severity. Arch. Neurol. 2004, 61, 902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Dalakas, M.C. The role of IVIg in the treatment of patients with stiff person syndrome and other neurological diseases associated with anti-GAD antibodies. J. Neurol. 2005, 252, I19–I25. [Google Scholar] [CrossRef] [PubMed]
  34. Dalakas, M.C.; Rakocevic, G.; Dambrosia, J.M.; Alexopoulos, H.; McElroy, B. A double-blind, placebo-controlled study of rituximab in patients with stiff person syndrome. Ann. Neurol. 2017, 82, 271–277. [Google Scholar] [CrossRef] [PubMed]
  35. Di Giacomo, R.; Deleo, F.; Pastori, C.; Didato, G.; Andreetta, F.; Del Sole, A.; de Curtis, M.; Villani, F. Predictive value of high titer of GAD65 antibodies in a case of limbic encephalitis. J. Neuroimmunol. 2019, 337, 577063. [Google Scholar] [CrossRef] [PubMed]
  36. Saiz, A.; Arpa, J.; Sagasta, A.; Casamitjana, R.; Zarranz, J.J.; Tolosa, E.; Graus, F. Autoantibodies to glutamic acid decarboxylase in three patients with cerebellar ataxia, late-onset insulin-dependent diabetes mellitus, and polyendocrine autoimmunity. Neurology 1997, 49, 1026–1030. [Google Scholar] [CrossRef]
  37. Höftberger, R. Neuroimmunology: An expanding frontier in autoimmunity. Front. Immunol. 2015, 6, 206. [Google Scholar] [CrossRef] [Green Version]
  38. Ricken, G.; Schwaiger, C.; De Simoni, D.; Pichler, V.; Lang, J.; Glatter, S.; Macher, S.; Rommer, P.S.; Scholze, P.; Kubista, H.; et al. Detection Methods for Autoantibodies in Suspected Autoimmune Encephalitis. Front. Neurol. 2018, 9, 841. [Google Scholar] [CrossRef] [Green Version]
  39. Solimena, M.; Folli, F.; Denis-Donini, S.; Comi, G.C.; Pozza, G.; De Camilli, P.; Vicari, A.M. Autoantibodies to glutamic acid decarboxylase in a patient with Stiff-Man syndrome, epilepsy, and type I diabetes mellitus. N. Engl. J. Med. 1988, 318, 1012–1020. [Google Scholar] [CrossRef]
  40. Pittock, S.J.; Yoshikawa, H.; Ahlskog, J.E.; Tisch, S.H.; Benarroch, E.E.; Kryzer, T.J.; Lennon, V.A. Glutamic acid decarboxylase autoimmunity with brainstem, extrapyramidal, and spinal cord dysfunction. Mayo Clin. Proc. 2006, 81, 1207–1214. [Google Scholar] [CrossRef] [Green Version]
  41. Oertel, W.H.; Schmechel, D.E.; Mugnaini, E.; Tappaz, M.L.; Kopin, I.J. Immunocytochemical localization of glutamate decarboxylase in rat cerebellum with a new antiserum. Neuroscience 1981, 6, 2715–2735. [Google Scholar] [CrossRef]
  42. Solimena, M.; Folli, F.; Aparisi, R.; Pozza, G.; De Camilli, P. Autoantibodies to GABA-ergic neurons and pancreatic beta cells in stiff-man syndrome. N. Engl. J. Med. 1990, 322, 1555–1560. [Google Scholar] [CrossRef] [PubMed]
  43. Aydin, S. A short history, principles, and types of ELISA, and our laboratory experience with peptide/protein analyses using ELISA. Peptides 2015, 72, 4–15. [Google Scholar] [CrossRef] [PubMed]
  44. Walikonis, J.E.; Lennon, V.A. Radioimmunoassay for glutamic acid decarboxylase (GAD65) autoantibodies as a diagnostic aid for stiff-man syndrome and a correlate of susceptibility to type 1 diabetes mellitus. Mayo Clin. Proc. 1998, 73, 1161–1166. [Google Scholar] [CrossRef] [PubMed]
  45. McKeon, A.; Robinson, M.T.; McEvoy, K.M.; Matsumoto, J.Y.; Lennon, V.A.; Ahlskog, J.E.; Pittock, S.J. Stiff-man syndrome and variants: Clinical course, treatments, and outcomes. Arch. Neurol. 2012, 69, 230–238. [Google Scholar] [CrossRef] [Green Version]
  46. Sivilotti, L.; Nistri, A. GABA receptor mechanisms in the central nervous system. Prog. Neurobiol. 1991, 36, 35–92. [Google Scholar] [CrossRef]
  47. DeFelipe, J.; López-Cruz, P.L.; Benavides-Piccione, R.; Bielza, C.; Larrañaga, P.; Anderson, S.; Burkhalter, A.; Cauli, B.; Fairén, A.; Feldmeyer, D.; et al. New insights into the classification and nomenclature of cortical GABAergic interneurons. Nat. Rev. Neurosci. 2013, 14, 202–216. [Google Scholar] [CrossRef] [Green Version]
  48. Frangaj, A.; Fan, Q.R. Structural biology of GABAB receptor. Neuropharmacology 2018, 136, 68–79. [Google Scholar] [CrossRef]
  49. Bowery, N.G.; Bettler, B.; Froestl, W.; Gallagher, J.P.; Marshall, F.; Raiteri, M.; Bonner, T.I.; Enna, S.J. International Union of Pharmacology. XXXIII. Mammalian gamma-aminobutyric acid(B) receptors: Structure and function. Pharmacol. Rev. 2002, 54, 247–264. [Google Scholar] [CrossRef]
  50. Mitoma, H.; Adhikari, K.; Aeschlimann, D.; Chattopadhyay, P.; Hadjivassiliou, M.; Hampe, C.S.; Honnorat, J.; Joubert, B.; Kakei, S.; Lee, J.; et al. Consensus Paper: Neuroimmune Mechanisms of Cerebellar Ataxias. Cerebellum 2016, 15, 213–232. [Google Scholar] [CrossRef] [Green Version]
  51. Hansen, N.; Grünewald, B.; Weishaupt, A.; Colaço, M.N.; Toyka, K.V.; Sommer, C.; Geis, C. Human Stiff person syndrome IgG-containing high-titer anti-GAD65 autoantibodies induce motor dysfunction in rats. Exp. Neurol. 2013, 239, 202–209. [Google Scholar] [CrossRef]
  52. Koerner, C.; Wieland, B.; Richter, W.; Meinck, H.-M. Stiff-person syndromes: Motor cortex hyperexcitability correlates with anti-GAD autoimmunity. Neurology 2004, 62, 1357–1362. [Google Scholar] [CrossRef] [PubMed]
  53. Tian, N.; Petersen, C.; Kash, S.; Baekkeskov, S.; Copenhagen, D.; Nicoll, R. The role of the synthetic enzyme GAD65 in the control of neuronal gamma-aminobutyric acid release. Proc. Natl. Acad. Sci. USA 1999, 96, 12911–12916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Mitoma, H.; Song, S.Y.; Ishida, K.; Yamakuni, T.; Kobayashi, T.; Mizusawa, H. Presynaptic impairment of cerebellar inhibitory synapses by an autoantibody to glutamate decarboxylase. J. Neurol. Sci. 2000, 175, 40–44. [Google Scholar] [CrossRef]
  55. Mitoma, H.; Ishida, K.; Shizuka-Ikeda, M.; Mizusawa, H. Dual impairment of GABAA- and GABAB-receptor-mediated synaptic responses by autoantibodies to glutamic acid decarboxylase. J. Neurol. Sci. 2003, 208, 51–56. [Google Scholar] [CrossRef]
  56. Bettler, B.; Kaupmann, K.; Mosbacher, J.; Gassmann, M. Molecular structure and physiological functions of GABA(B) receptors. Physiol. Rev. 2004, 84, 835–867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Terunuma, M. Diversity of structure and function of GABAB receptors: A complexity of GABAB-mediated signaling. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2018, 94, 390–411. [Google Scholar] [CrossRef] [Green Version]
  58. Levy, L.M.; Dalakas, M.C.; Floeter, M.K. The stiff-person syndrome: An autoimmune disorder affecting neurotransmission of gamma-aminobutyric acid. Ann. Intern. Med. 1999, 131, 522–530. [Google Scholar] [CrossRef] [Green Version]
  59. Mitoma, H.; Manto, M.; Hampe, C.S. Pathogenic Roles of Glutamic Acid Decarboxylase 65 Autoantibodies in Cerebellar Ataxias. J. Immunol. Res. 2017, 2017, 2913297. [Google Scholar] [CrossRef]
  60. Takenoshita, H.; Shizuka-Ikeda, M.; Mitoma, H.; Song, S.; Harigaya, Y.; Igeta, Y.; Yaguchi, M.; Ishida, K.; Shoji, M.; Tanaka, M.; et al. Presynaptic inhibition of cerebellar GABAergic transmission by glutamate decarboxylase autoantibodies in progressive cerebellar ataxia. J. Neurol. Neurosurg. Psychiatry 2001, 70, 386–389. [Google Scholar] [CrossRef] [Green Version]
  61. Ishida, K.; Mitoma, H.; Song, S.Y.; Uchihara, T.; Inaba, A.; Eguchi, S.; Kobayashi, T.; Mizusawa, H. Selective suppression of cerebellar GABAergic transmission by an autoantibody to glutamic acid decarboxylase. Ann. Neurol. 1999, 46, 263–267. [Google Scholar] [CrossRef]
  62. Ishida, K.; Mitoma, H.; Mizusawa, H. Reversibility of cerebellar GABAergic synapse impairment induced by anti-glutamic acid decarboxylase autoantibodies. J. Neurol. Sci. 2008, 271, 186–190. [Google Scholar] [CrossRef]
  63. Manto, M.-U.; Laute, M.-A.; Aguera, M.; Rogemond, V.; Pandolfo, M.; Honnorat, J. Effects of anti-glutamic acid decarboxylase antibodies associated with neurological diseases: GAD-Ab and Cerebellar Function. Ann. Neurol. 2007, 61, 544–551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Manto, M.; Mitoma, H.; Hampe, C.S. Anti-GAD Antibodies and the Cerebellum: Where Do We Stand? Cerebellum 2019, 18, 153–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ishikawa, T.; Tomatsu, S.; Tsunoda, Y.; Lee, J.; Hoffman, D.S.; Kakei, S. Releasing dentate nucleus cells from Purkinje cell inhibition generates output from the cerebrocerebellum. PLoS ONE 2014, 9, e108774. [Google Scholar] [CrossRef] [Green Version]
  66. Vianello, M.; Bisson, G.; Dal Maschio, M.; Vassanelli, S.; Girardi, S.; Mucignat, C.; Fountzoulas, K.; Giometto, B. Increased spontaneous activity of a network of hippocampal neurons in culture caused by suppression of inhibitory potentials mediated by anti-gad antibodies. Autoimmunity 2008, 41, 66–73. [Google Scholar] [CrossRef] [PubMed]
  67. Ziegler, A.-G.; Nepom, G.T. Prediction and Pathogenesis in Type 1 Diabetes. Immunity 2010, 32, 468–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Arneth, B. Activation of CD4+ and CD8+ T-lymphocytes by insulin and GAD in patients with type 1 or 2 diabetes mellitus. Endocr. Connect. 2017, 6, 758–765. [Google Scholar] [CrossRef] [Green Version]
  69. Mallone, R.; Martinuzzi, E.; Blancou, P.; Novelli, G.; Afonso, G.; Dolz, M.; Bruno, G.; Chaillous, L.; Chatenoud, L.; Bach, J.-M.; et al. CD8+ T-cell responses identify beta-cell autoimmunity in human type 1 diabetes. Diabetes 2007, 56, 613–621. [Google Scholar] [CrossRef]
  70. McKeon, A.; Pittock, S.J. Paraneoplastic encephalomyelopathies: Pathology and mechanisms. Acta Neuropathol. 2011, 122, 381–400. [Google Scholar] [CrossRef]
  71. Burton, A.R.; Baquet, Z.; Eisenbarth, G.S.; Tisch, R.; Smeyne, R.; Workman, C.J.; Vignali, D.A.A. Central Nervous System Destruction Mediated by Glutamic Acid Decarboxylase-Specific CD4+ T Cells. J. Immunol. 2010, 184, 4863–4870. [Google Scholar] [CrossRef] [Green Version]
  72. Ishizawa, K.; Komori, T.; Okayama, K.; Qin, X.; Kaneko, K.; Sasaki, S.; Iwata, M. Large motor neuron involvement in Stiff-man syndrome: A qualitative and quantitative study. Acta Neuropathol. 1999, 97, 63–70. [Google Scholar] [CrossRef] [PubMed]
  73. Warren, J.D.; Scott, G.; Blumbergs, P.C.; Thompson, P.D. Pathological evidence of encephalomyelitis in the stiff man syndrome with anti-GAD antibodies. J. Clin. Neurosci. 2002, 9, 328–329. [Google Scholar] [CrossRef] [PubMed]
  74. Holmøy, T.; Skorstad, G.; Røste, L.S.; Scheie, D.; Alvik, K. Stiff person syndrome associated with lower motor neuron disease and infiltration of cytotoxic T cells in the spinal cord. Clin. Neurol. Neurosurg. 2009, 111, 708–712. [Google Scholar] [CrossRef] [PubMed]
  75. Ishida, K.; Mitoma, H.; Wada, Y.; Oka, T.; Shibahara, J.; Saito, Y.; Murayama, S.; Mizusawa, H. Selective loss of Purkinje cells in a patient with anti-glutamic acid decarboxylase antibody-associated cerebellar ataxia. J. Neurol. Neurosurg. Psychiatry 2007, 78, 190–192. [Google Scholar] [CrossRef] [Green Version]
  76. Bien, C.G.; Vincent, A.; Barnett, M.H.; Becker, A.J.; Blümcke, I.; Graus, F.; Jellinger, K.A.; Reuss, D.E.; Ribalta, T.; Schlegel, J.; et al. Immunopathology of autoantibody-associated encephalitides: Clues for pathogenesis. Brain 2012, 135, 1622–1638. [Google Scholar] [CrossRef] [Green Version]
  77. Espay, A.J.; Chen, R. Rigidity and spasms from autoimmune encephalomyelopathies: Stiff-person syndrome. Muscle Nerve 2006, 34, 677–690. [Google Scholar] [CrossRef]
  78. Dubey, D.; Pittock, S.J.; Kelly, C.R.; McKeon, A.; Lopez-Chiriboga, A.S.; Lennon, V.; Gadoth, A.; Smith, C.Y.; Bryant, S.C.; Klein, C.J.; et al. Autoimmune Encephalitis Epidemiology and a comparison to Infectious Encephalitis. Ann. Neurol. 2018, 83, 166–177. [Google Scholar] [CrossRef]
  79. Dalakas, M.C.; Fujii, M.; Li, M.; McElroy, B. The clinical spectrum of anti-GAD antibody-positive patients with stiff-person syndrome. Neurology 2000, 55, 1531–1535. [Google Scholar] [CrossRef] [Green Version]
  80. Hadjivassiliou, M.; Martindale, J.; Shanmugarajah, P.; Grünewald, R.A.; Sarrigiannis, P.G.; Beauchamp, N.; Garrard, K.; Warburton, R.; Sanders, D.S.; Friend, D.; et al. Causes of progressive cerebellar ataxia: Prospective evaluation of 1500 patients. J. Neurol. Neurosurg. Psychiatry 2017, 88, 301–309. [Google Scholar] [CrossRef]
  81. Nanri, K.; Okuma, M.; Sato, S.; Yoneda, M.; Taguchi, T.; Mitoma, H.; Yamada, J.; Unezaki, S.; Nagatani, T.; Otsubo, S.; et al. Prevalence of Autoantibodies and the Efficacy of Immunotherapy for Autoimmune Cerebellar Ataxia. Intern. Med. 2016, 55, 449–454. [Google Scholar] [CrossRef] [Green Version]
  82. Brenner, T.; Sills, G.J.; Hart, Y.; Howell, S.; Waters, P.; Brodie, M.J.; Vincent, A.; Lang, B. Prevalence of neurologic autoantibodies in cohorts of patients with new and established epilepsy. Epilepsia 2013, 54, 1028–1035. [Google Scholar] [CrossRef] [PubMed]
  83. Lilleker, J.B.; Biswas, V.; Mohanraj, R. Glutamic acid decarboxylase (GAD) antibodies in epilepsy: Diagnostic yield and therapeutic implications. Seizure 2014, 23, 598–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Errichiello, L.; Perruolo, G.; Pascarella, A.; Formisano, P.; Minetti, C.; Striano, S.; Zara, F.; Striano, P. Autoantibodies to glutamic acid decarboxylase (GAD) in focal and generalized epilepsy: A study on 233 patients. J. Neuroimmunol. 2009, 211, 120–123. [Google Scholar] [CrossRef] [PubMed]
  85. Bien, C.G.; Scheffer, I.E. Autoantibodies and epilepsy. Epilepsia 2011, 52, 18–22. [Google Scholar] [CrossRef]
  86. Sørgjerd, E.P.; Thorsby, P.M.; Torjesen, P.A.; Skorpen, F.; Kvaløy, K.; Grill, V. Presence of anti-GAD in a non-diabetic population of adults; time dynamics and clinical influence: Results from the HUNT study. BMJ Open Diabetes Res. Care 2015, 3, e000076. [Google Scholar] [CrossRef] [Green Version]
  87. Honnorat, J.; Saiz, A.; Giometto, B.; Vincent, A.; Brieva, L.; de Andres, C.; Maestre, J.; Fabien, N.; Vighetto, A.; Casamitjana, R.; et al. Cerebellar ataxia with anti-glutamic acid decarboxylase antibodies: Study of 14 patients. Arch. Neurol. 2001, 58, 225–230. [Google Scholar] [CrossRef] [Green Version]
  88. Liimatainen, S.; Peltola, M.; Sabater, L.; Fallah, M.; Kharazmi, E.; Haapala, A.-M.; Dastidar, P.; Knip, M.; Saiz, A.; Peltola, J. Clinical significance of glutamic acid decarboxylase antibodies in patients with epilepsy. Epilepsia 2010, 51, 760–767. [Google Scholar] [CrossRef]
  89. Muñiz-Castrillo, S.; Ambati, A.; Dubois, V.; Vogrig, A.; Joubert, B.; Rogemond, V.; Picard, G.; Lin, L.; Fabien, N.; Mignot, E.; et al. Primary DQ effect in the association between HLA and neurological syndromes with anti-GAD65 antibodies. J. Neurol. 2020. [Google Scholar] [CrossRef]
  90. Muñiz-Castrillo, S.; Vogrig, A.; Honnorat, J. Associations between HLA and autoimmune neurological diseases with autoantibodies. Autoimmun. Highlights 2020, 11, 2. [Google Scholar] [CrossRef]
  91. Gough, S.C.L.; Simmonds, M.J. The HLA Region and Autoimmune Disease: Associations and Mechanisms of Action. Curr. Genom. 2007, 8, 453–465. [Google Scholar] [CrossRef] [Green Version]
  92. Pugliese, A.; Solimena, M.; Awdeh, Z.L.; Alper, C.A.; Bugawan, T.; Erlich, H.A.; De Camilli, P.; Eisenbarth, G.S. Association of HLA-DQB1*0201 with stiff-man syndrome. J. Clin. Endocrinol. Metab. 1993, 77, 1550–1553. [Google Scholar] [CrossRef] [PubMed]
  93. Joubert, B.; Belbezier, A.; Haesebaert, J.; Rheims, S.; Ducray, F.; Picard, G.; Rogemond, V.; Psimaras, D.; Berzero, G.; Desestret, V.; et al. Long-term outcomes in temporal lobe epilepsy with glutamate decarboxylase antibodies. J. Neurol. 2020. [Google Scholar] [CrossRef] [PubMed]
  94. Thomas, S.; Critchley, P.; Lawden, M.; Farooq, S.; Thomas, A.; Proudlock, F.A.; Constantinescu, C.S.; Gottlob, I. Stiff person syndrome with eye movement abnormality, myasthenia gravis, and thymoma. J. Neurol. Neurosurg. Psychiatry 2005, 76, 141–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Tanaka, H.; Matsumura, A.; Okumura, M.; Kitaguchi, M.; Yamamoto, S.; Iuchi, K. Stiff man syndrome with thymoma. Ann. Thorac. Surg. 2005, 80, 739–741. [Google Scholar] [CrossRef]
  96. Rosin, L.; DeCamilli, P.; Butler, M.; Solimena, M.; Schmitt, H.-P.; Morgenthaler, N.; Meinck, H.-M. Stiff-man syndrome in a woman with breast cancer: An uncommon central nervous system paraneoplastic syndrome. Neurology 1998, 50, 94–98. [Google Scholar] [CrossRef]
  97. Silverman, I.E. Paraneoplastic stiff limb syndrome. J. Neurol. Neurosurg. Psychiatry 1999, 67, 126–127. [Google Scholar] [CrossRef] [Green Version]
  98. Ariño, H.; Höftberger, R.; Gresa-Arribas, N.; Martínez-Hernandez, E.; Armangue, T.; Kruer, M.C.; Arpa, J.; Domingo, J.; Rojc, B.; Bataller, L.; et al. Paraneoplastic Neurological Syndromes and Glutamic Acid Decarboxylase Antibodies. JAMA Neurol. 2015, 72, 874–881. [Google Scholar] [CrossRef]
  99. Bien, C.G. Association of Paraneoplastic Neurological Disorders With Glutamic Acid Decarboxylase Antibodies. JAMA Neurol. 2015, 72, 861–862. [Google Scholar] [CrossRef]
  100. Hernández-Echebarría, L.; Saiz, A.; Arés, A.; Tejada, J.; García-Tuñón, L.; Nieves, C.; Graus, F. Paraneoplastic encephalomyelitis associated with pancreatic tumor and anti-GAD antibodies. Neurology 2006, 66, 450–451. [Google Scholar] [CrossRef]
  101. Bataller, L.; Valero, C.; Díaz, R.; Froufe, A.; Garcia-Zarza, A.; Ribalta, T.; Vilchez, J.J.; Saiz, A. Cerebellar ataxia associated with neuroendocrine thymic carcinoma and GAD antibodies. J. Neurol. Neurosurg. Psychiatry 2009, 80, 696–697. [Google Scholar] [CrossRef]
  102. Graus, F.; Dalmau, J. Paraneoplastic neurological syndromes in the era of immune-checkpoint inhibitors. Nat. Rev. Clin. Oncol. 2019, 16, 535–548. [Google Scholar] [CrossRef] [PubMed]
  103. Shah, S.; Dunn-Pirio, A.; Luedke, M.; Morgenlander, J.; Skeen, M.; Eckstein, C. Nivolumab-Induced Autoimmune Encephalitis in Two Patients with Lung Adenocarcinoma. Case Rep. Neurol. Med. 2018, 2018, 2548528. [Google Scholar] [CrossRef] [PubMed]
  104. Chung, M.; Jaffer, M.; Verma, N.; Mokhtari, S.; Ramsakal, A.; Peguero, E. Immune checkpoint inhibitor induced anti-glutamic acid decarboxylase 65 (Anti-GAD 65) limbic encephalitis responsive to intravenous immunoglobulin and plasma exchange. J. Neurol. 2019, 1. [Google Scholar] [CrossRef] [PubMed]
  105. Lee, S.; Morgan, A.; Shah, S.; Ebeling, P.R. Rapid-onset diabetic ketoacidosis secondary to nivolumab therapy. Endocrinol. Diabetes Metab. Case Rep. 2018, 2018. [Google Scholar] [CrossRef] [PubMed]
  106. De Filette, J.M.K.; Pen, J.J.; Decoster, L.; Vissers, T.; Bravenboer, B.; Van der Auwera, B.J.; Gorus, F.K.; Roep, B.O.; Aspeslagh, S.; Neyns, B.; et al. Immune checkpoint inhibitors and type 1 diabetes mellitus: A case report and systematic review. Eur. J. Endocrinol. 2019, 181, 363–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Tzoulis, P.; Corbett, R.W.; Ponnampalam, S.; Baker, E.; Heaton, D.; Doulgeraki, T.; Stebbing, J. Nivolumab-induced fulminant diabetic ketoacidosis followed by thyroiditis. Endocrinol. Diabetes Metab. Case Rep. 2018, 2018. [Google Scholar] [CrossRef] [Green Version]
  108. Moersch, F.P.; Woltman, H.W. Progressive fluctuating muscular rigidity and spasm (“stiff-man” syndrome); report of a case and some observations in 13 other cases. Proc. Staff Meet. Mayo Clin. 1956, 31, 421–427. [Google Scholar]
  109. Barker, R.; Revesz, T.; Thom, M.; Marsden, C.; Brown, P. Review of 23 patients affected by the stiff man syndrome: Clinical subdivision into stiff trunk (man) syndrome, stiff limb syndrome, and progressive encephalomyelitis with rigidity. J. Neurol. Neurosurg. Psychiatry 1998, 65, 633–640. [Google Scholar] [CrossRef] [Green Version]
  110. Meinck, H.M.; Ricker, K.; Conrad, B. The stiff-man syndrome: New pathophysiological aspects from abnormal exteroceptive reflexes and the response to clomipramine, clonidine, and tizanidine. J. Neurol. Neurosurg. Psychiatry 1984, 47, 280–287. [Google Scholar] [CrossRef]
  111. Mitoma, H.; Manto, M.; Hampe, C.S. Immune-mediated Cerebellar Ataxias: Practical Guidelines and Therapeutic Challenges. Curr. Neuropharmacol. 2019, 17, 33–58. [Google Scholar] [CrossRef]
  112. Aguiar, T.S.; Fragoso, A.; de Albuquerque, C.R.; de Teixeira, P.F.; de Souza, M.V.L.; Zajdenverg, L.; Alves-Leon, S.V.; Rodacki, M.; Lima, M.A.S.D. de Clinical characteristics of patients with cerebellar ataxia associated with anti-GAD antibodies. Arq. Neuropsiquiatr. 2017, 75, 142–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Tripathi, M.; Tripathi, M.; Roy, S.G.; Parida, G.K.; Ihtisham, K.; Dash, D.; Damle, N.; Shamim, S.A.; Bal, C. Metabolic topography of autoimmune non-paraneoplastic encephalitis. Neuroradiology 2018, 60, 189–198. [Google Scholar] [CrossRef] [PubMed]
  114. Graus, F.; Delattre, J.Y.; Antoine, J.C.; Dalmau, J.; Giometto, B.; Grisold, W.; Honnorat, J.; Smitt, P.S.; Vedeler, C.; Verschuuren, J.J.G.M.; et al. Recommended diagnostic criteria for paraneoplastic neurological syndromes. J. Neurol. Neurosurg. Psychiatry 2004, 75, 1135–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Kanter, I.C.; Huttner, H.B.; Staykov, D.; Biermann, T.; Struffert, T.; Kerling, F.; Hilz, M.-J.; Schellinger, P.D.; Schwab, S.; Bardutzky, J. Cyclophosphamide for anti-GAD antibody-positive refractory status epilepticus. Epilepsia 2008, 49, 914–920. [Google Scholar] [CrossRef]
  116. Triplett, J.; Vijayan, S.; MacDonald, A.; Lawn, N.; McLean-Tooke, A.; Bynevelt, M.; Phatouros, C.; Chemmanam, T. Fulminant Anti-GAD antibody encephalitis presenting with status epilepticus requiring aggressive immunosuppression. J. Neuroimmunol. 2018, 323, 119–124. [Google Scholar] [CrossRef]
  117. Rakocevic, G.; Raju, R.; Semino-Mora, C.; Dalakas, M.C. Stiff person syndrome with cerebellar disease and high-titer anti-GAD antibodies. Neurology 2006, 67, 1068–1070. [Google Scholar] [CrossRef]
  118. Jazebi, N.; Rodrigo, S.; Gogia, B.; Shawagfeh, A. Anti-glutamic acid decarboxylase (GAD) positive cerebellar Ataxia with transitioning to progressive encephalomyelitis with rigidity and myoclonus (PERM), responsive to immunotherapy: A case report and review of literature. J. Neuroimmunol. 2019, 332, 135–137. [Google Scholar] [CrossRef]
  119. Wirth, T.; Kaeuffer, C.; Chanson, J.B.; Echaniz-Laguna, A.; Renaud, M.; Anheim, M.; Schneider, F.; Tranchant, C. Progressive encephalomyelitis with rigidity and myoclonus, a diagnostic challenge. Rev. Neurol. (Paris) 2018, 174, 343–346. [Google Scholar] [CrossRef]
  120. Joubert, B.; Rostásy, K.; Honnorat, J. Immune-mediated ataxias. Handb. Clin. Neurol. 2018, 155, 313–332. [Google Scholar] [CrossRef]
  121. Nemni, R.; Braghi, S.; Natali-Sora, M.G.; Lampasona, V.; Bonifacio, E.; Comi, G.; Canal, N. Autoantibodies to glutamic acid decarboxylase in palatal myoclonus and epilepsy. Ann. Neurol. 1994, 36, 665–667. [Google Scholar] [CrossRef]
  122. Vianello, M.; Morello, F.; Scaravilli, T.; Tavolato, B.; Giometto, B. Tremor of the mouth floor and anti-glutamic acid decarboxylase autoantibodies. Eur. J. Neurol. 2003, 10, 513–514. [Google Scholar] [CrossRef]
  123. Liu, Z.; Huang, Q.; Li, H.; Qiu, W.; Chen, B.; Luo, J.; Yang, H.; Liu, T.; Liu, S.; Xu, H.; et al. Glutamic Acid Decarboxylase Antibody in a Patient with Myelitis: A Retrospective Study. Neuroimmunomodulation 2018, 25, 68–72. [Google Scholar] [CrossRef]
  124. Fileccia, E.; Rinaldi, R.; Liguori, R.; Incensi, A.; D’Angelo, R.; Giannoccaro, M.P.; Donadio, V. Post-ganglionic autonomic neuropathy associated with anti-glutamic acid decarboxylase antibodies. Clin. Auton. Res. 2017, 27, 51–55. [Google Scholar] [CrossRef]
  125. Chattopadhyay, S.; Kriscenski-Perry, E.; Wenger, D.A.; Pearce, D.A. An autoantibody to GAD65 in sera of patients with juvenile neuronal ceroid lipofuscinoses. Neurology 2002, 59, 1816–1817. [Google Scholar] [CrossRef] [PubMed]
  126. Piccolo, G.; Cosi, V.; Zandrini, C.; Moglia, A. Steroid-responsive and dependent stiff-man syndrome: A clinical and electrophysiological study of two cases. Ital. J. Neurol. Sci. 1988, 9, 559–566. [Google Scholar] [CrossRef] [PubMed]
  127. Brashear, H.R.; Phillips, L.H. Autoantibodies to GABAergic neurons and response to plasmapheresis in stiff-man syndrome. Neurology 1991, 41, 1588–1592. [Google Scholar] [CrossRef] [PubMed]
  128. Sevy, A.; Franques, J.; Chiche, L.; Pouget, J.; Attarian, S. Successful treatment with rituximab in a refractory Stiff-person syndrome. Rev. Neurol. (Paris) 2012, 168, 375–378. [Google Scholar] [CrossRef] [PubMed]
  129. Bacorro, E.; Tehrani, R. Stiff-Person Syndrome Persistent Elevation of Glutamic Acid Decarboxylase Antibodies Despite Successful Treatment with Rituximab. J. Clin. Rheumatol. Pract. Rep. Rheum. Musculoskelet. Dis. 2010, 16, 237–239. [Google Scholar] [CrossRef]
  130. Pedroso, J.L.; Braga-Neto, P.; Dutra, L.A.; Barsottini, O.G.P. Cerebellar ataxia associated to anti-glutamic acid decarboxylase autoantibody (anti-GAD): Partial improvement with intravenous immunoglobulin therapy. Arq. Neuropsiquiatr. 2011, 69, 993. [Google Scholar] [CrossRef] [Green Version]
  131. Abele, M.; Weller, M.; Mescheriakov, S.; Bürk, K.; Dichgans, J.; Klockgether, T. Cerebellar ataxia with glutamic acid decarboxylase autoantibodies. Neurology 1999, 52, 857–859. [Google Scholar] [CrossRef]
  132. Lauria, G.; Pareyson, D.; Pitzolu, M.G.; Bazzigaluppi, E. Excellent response to steroid treatment in anti-GAD cerebellar ataxia. Lancet Neurol. 2003, 2, 634–635. [Google Scholar] [CrossRef]
  133. Virgilio, R.; Corti, S.; Agazzi, P.; Santoro, D.; Lanfranconi, S.; Candelise, L.; Bresolin, N.; Comi, G.P.; Bersano, A. Effect of steroid treatment in cerebellar ataxia associated with anti-glutamic acid decarboxylase antibodies. J. Neurol. Neurosurg. Psychiatry 2009, 80, 95–96. [Google Scholar] [CrossRef] [PubMed]
  134. Kuchling, J.; Shababi-Klein, J.; Nümann, A.; Gerischer, L.M.; Harms, L.; Prüss, H. GAD Antibody-Associated Late-Onset Cerebellar Ataxia in Two Female Siblings. Case Rep. Neurol. 2014, 6, 264–270. [Google Scholar] [CrossRef] [PubMed]
  135. Marchiori, G.C.; Vaglia, A.; Vianello, M.; Bardin, P.G.; Giometto, B. Encephalitis associated with glutamic acid decarboxylase autoantibodies. Neurology 2001, 56, 814. [Google Scholar] [CrossRef]
  136. Mazzi, G.; Roia, D.D.; Cruciatti, B.; Matà, S.; Catapano, R. Plasma exchange for anti-GAD associated non paraneoplastic limbic encephalitis. Transfus. Apher. Sci. 2008, 39, 229–233. [Google Scholar] [CrossRef]
  137. Malter, M.P.; Frisch, C.; Zeitler, H.; Surges, R.; Urbach, H.; Helmstaedter, C.; Elger, C.E.; Bien, C.G. Treatment of immune-mediated temporal lobe epilepsy with GAD antibodies. Seizure 2015, 30, 57–63. [Google Scholar] [CrossRef] [Green Version]
  138. Hao, W.; Davis, C.; Hirsch, I.B.; Eng, L.J.; Daniels, T.; Walsh, D.; Lernmark, A. Plasmapheresis and immunosuppression in stiff-man syndrome with type 1 diabetes: A 2-year study. J. Neurol. 1999, 246, 731–735. [Google Scholar] [CrossRef]
  139. Fekete, R.; Jankovic, J. Childhood Stiff-Person syndrome improved with rituximab. Case Rep. Neurol. 2012, 4, 92–96. [Google Scholar] [CrossRef]
  140. Mitoma, H.; Hadjivassiliou, M.; Honnorat, J. Guidelines for treatment of immune-mediated cerebellar ataxias. Cerebellum Ataxias 2015, 2, 14. [Google Scholar] [CrossRef] [Green Version]
  141. Hansen, N.; Widman, G.; Witt, J.-A.; Wagner, J.; Becker, A.J.; Elger, C.E.; Helmstaedter, C. Seizure control and cognitive improvement via immunotherapy in late onset epilepsy patients with paraneoplastic versus GAD65 autoantibody-associated limbic encephalitis. Epilepsy Behav. 2016, 65, 18–24. [Google Scholar] [CrossRef]
Figure 1. The structural coupling between gamma-aminobutyric acid (GABA) synthesis and vesicular GABA transport into a synaptic vesicle (SV). GAD65 is anchored to SVs through a protein complex with the chaperone HSC70, followed by association of HSC70-GAD65 complex to Cysteine-String Protein (CSP), Vesicular GABA transporter (VGAT) and Calcium/calmoduline protein kinase (CaMKII) on SVs. The numbers indicate the different required steps.
Figure 1. The structural coupling between gamma-aminobutyric acid (GABA) synthesis and vesicular GABA transport into a synaptic vesicle (SV). GAD65 is anchored to SVs through a protein complex with the chaperone HSC70, followed by association of HSC70-GAD65 complex to Cysteine-String Protein (CSP), Vesicular GABA transporter (VGAT) and Calcium/calmoduline protein kinase (CaMKII) on SVs. The numbers indicate the different required steps.
Ijms 21 03701 g001
Table 1. Clinical and paraclinical characteristics of SPS, CA, and LE associated with GAD Ab in the main series reported in literature.
Table 1. Clinical and paraclinical characteristics of SPS, CA, and LE associated with GAD Ab in the main series reported in literature.
ReferenceNumber of PatientsMedian Age (Range)Female GenderAssociated Autoimmune DisordersParaneoplastic CasesNeurological Symptoms/PhenotypesIntrathecal Synthesis of GAD AbOligoclonal Bands in the CSF
SPS
Saiz et al., 2008 [28]2256 (14–77)19/22 (86%)T1DM (59%), thyroiditis (18%), vitiligo (9%)noneclassic SPS (82%), focal SPS (18%)11/13 (85%)5/14 (35%)
McKeon et al., 2012 [45]79NANAT1DM (43%), thyroiditis (35%), vitiligo (9%), PA (8%)3/79 (4%); thyroid, kidney, colonclassic SPS (75%), focal SPS (24%), PERM (1%)NANA
Arino et al., 2014 [18]2856 (19–77)26/28 (93%)T1DM (50%), thyroiditis (25%), PA (11%), vitiligo (11%)1/28 (4%) **; breast cancerNA9/11 (82%)5/17 (29%)
Gresa-Arribas et al., 2015 [15]3253 (5–77)29/32 (91%)T1DM (48%), thyroiditis (28%), other (16%)excluded from the studyNANA4/15 (27%)
Cerebellar Ataxias
Honnorat et al., 2001 [87]1451 (20–74)1314 (93%)T1DM (71%), thyroiditis (57%), PA (14%), myasthenia gravis (7%)2/14 (14%); thymomasgait ataxia (100%), limb ataxia (86%), nystagmus (86%), dysarthria (57%)5/6 (83%)10/14 (71%)
Saiz et al., 2008 [28]1759 (39–77)16/17 (94%)T1DM (53%), thyroiditis (41%), PA (12%), vitiligo (6%)2/17 (12%); NSCLC, neuroendocrine thymic carcinomagait ataxia (100%), limb ataxia (59%), dysarthria (65%), nystagmus (65%)12/12 (100%)9/13 (69%)
Arino et al., 2014 [18]3458 (33–80)28/34 (82%)T1DM (38%), thyroiditis (53%), PA (21%), vitiligo (6%)4/34 (12%) *; thymoma, endometrial carcinoma, breast cancer, MDSgait ataxia (91%), limb ataxia (74%), dysarthria (71%), nystagmus (59%)13/15 (87%)16/22 (73%)
Gresa-Arribas et al., 2015 [15]3960 (32–79)32/39 (82%)T1DM (38%), thyroiditis (60%), other (23%)excluded from the studyNANA18/24 (75%)
Limbic Encephalitis
Malter et al., 2010 [29]923 (17–66)7/9 (78%)T1DM (22%)noneseizures (100%), overt cognitive impairment or psychiatric disturbances (11%)9/9 (100%)5/8 (63%)
Gresa-Arribas et al., 2015 [15]1726 (12–49)12/15 (80%)T1DM (33%), thyroiditis (60%), other (12%)excluded from the studyNANA7/7 (100%)
Joubert et al., 2020 [93] 1530 (2–63)14/15(93%) Autoimmune diseases (60%)noneSeizures (53%), acute amnesia (67%), behavioral disorders (33%)NA12/14 (86%)
* Three tumors out of the four were diagnosed at least 7 years before the onset of CA, and the remaining one 6 years after the onset of CA. ** Cancer was diagnosed 20 years after the onset of SPS.
Table 2. Response to different immunotherapies in the main series of SPS, cerebellar ataxia, and limbic encephalitis associated with GAD Ab reported in literature.
Table 2. Response to different immunotherapies in the main series of SPS, cerebellar ataxia, and limbic encephalitis associated with GAD Ab reported in literature.
Number of PatientsTreatment ScheduleOutcome after TreatmentTreatment-Related ComplicationsMortality during FUReference
IVIg
Placebo controlled study
162 g/kg, divided into two daily doses, every month for 3 months11 out of 14 patients (79%) improve with regard to muscle rigidity, spasms, and functional ability to walkNone12.5%Dalakas et al., 2005 [33,23]
High-dose CS2Oral or intravenous steroids- Distal stiffness: 1/5 slight improvement and 1/5 worsening
- Axial stiffness: ½ slight improvement
NANABarker et al., 1998 [109]
High-dose CS2Prednisone 100 mg/d, and decrease1. improvement: no symptoms at 10 d
2. improvement
1. insomnia, increased anxiety, de- pressed mood
2. hypokalemia, cushingoid features
0%Piccolo et al., 1988 [126]
PE1No detailsStable: mRS 4NANAMcKeon et al., 2012 [45]
PE3No detailsStableNANABarker et al., 1998 [109]
PE1No detailsMarked improvementNANABrashear et al., 1991 [127]
Rituximab Double blind placebo-controlled study142 biweekly infusions of 1g eachPrimary outcome (change in stiffness scores at 6 months): non-significant effectSome infusion-related reactionsNADalakas et al., 2017 [34]
Rituximab1- 1000 mg at 0 and 14 dayPartial Improvement on scores:
-stiff: 4/6 to 1
- sensitivity: 5/7 to 4
NA0%Sevy et al., 2012 [128]
Rituximab11000 mg at 0 and 7 dayImprovement: mRS 4 to 1NA0%Bacorro et al. 2010 [129]
IVIg12 g/kg over 5 days every month for 3 monthsPartial improvement
(ICARS 65 to 37)
NANAPedroso et al., 2011 [130]
IVIg1IVIg every month for 2 monthsPartial improvement
(ICARS 59 to 48)
NA0%Abele et al., 1999 [131]
IVIg30.4 g/kg/day for 5 days, followed by two cycles of single monthly doses 1g/kgPartial improvement for 1/3
No improvement for 2/3
NANAAguiar et al., 2017 [112]
High-dose CS1MP 1000 mg/ day for 5 dImprovement: ICARS 60 to 36NA0%Lauria et al., 2003 [132]
High-dose CS1MP 1000 mg/day for 5 dImprovement: ICARS 38 to 22 at the beginning and ICARS 7 at 3 monthsNA0%Virgilio et al., 2009 [133]
PE + Rituximab27–10 cycles of plasmapheresis + 1000 mg rituximab IV1. High response during 1 month
2. No change
NA0%Kuchling et al., 2014 [134]
High-dose CS1MP 500 mg/d, 6 daysHigh improvementNA0%Marchiori et al., 2001 [135]
PE1Two cycles of 7 PE + 500 mg MPx3/AZA+ oral corticosteroidsImportant improvement the first month: decreasing of seizures, low response afterNANAMazzi et al., 2008 [136]
High-dose CS11median total dose 19 g (3–30 g)45% of response55%:
-Cushing syndrome in three patients,
hyperglycaemia, sleep disorders, nervousness and psychosis
NAMalter et al., 2015 [137]
IVIg5Dose: range 3–4 g for a median of 3 months20% (1 patient) with seizure response0%NAMalter et al., 2015 [137]
PE81 (or 2) sequence of 16 sessions in median (range: 11–26)13% (1 patient) of response0%NAMalter et al., 2015 [137]

Share and Cite

MDPI and ACS Style

Dade, M.; Berzero, G.; Izquierdo, C.; Giry, M.; Benazra, M.; Delattre, J.-Y.; Psimaras, D.; Alentorn, A. Neurological Syndromes Associated with Anti-GAD Antibodies. Int. J. Mol. Sci. 2020, 21, 3701. https://doi.org/10.3390/ijms21103701

AMA Style

Dade M, Berzero G, Izquierdo C, Giry M, Benazra M, Delattre J-Y, Psimaras D, Alentorn A. Neurological Syndromes Associated with Anti-GAD Antibodies. International Journal of Molecular Sciences. 2020; 21(10):3701. https://doi.org/10.3390/ijms21103701

Chicago/Turabian Style

Dade, Maëlle, Giulia Berzero, Cristina Izquierdo, Marine Giry, Marion Benazra, Jean-Yves Delattre, Dimitri Psimaras, and Agusti Alentorn. 2020. "Neurological Syndromes Associated with Anti-GAD Antibodies" International Journal of Molecular Sciences 21, no. 10: 3701. https://doi.org/10.3390/ijms21103701

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop