Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jul 7.
Published in final edited form as: Cell. 2022 Jul 7;185(14):2469–2477.e13. doi: 10.1016/j.cell.2022.06.025

Structural mechanisms of GABAA receptor autoimmune encephalitis

Colleen M Noviello 1, Jakob Kreye 2, Jinfeng Teng 1, Harald Prüss 3,#, Ryan E Hibbs 1,#
PMCID: PMC9394431  NIHMSID: NIHMS1818220  PMID: 35803245

Summary

Autoantibodies targeting neuronal membrane proteins can cause encephalitis, seizures, and severe behavioral abnormalities. While antibodies for several neuronal targets have been identified, structural details on how they regulate function are unknown. Here we determined cryo-electron microscopy structures of antibodies derived from an encephalitis patient bound to the γ-aminobutyric acid type A (GABAA) receptor. These antibodies induced a severe encephalitis by directly inhibiting GABAA function, resulting in nervous system hyperexcitability. The structures reveal mechanisms of GABAA inhibition and pathology. One antibody directly competes with neurotransmitter and locks the receptor in a resting-like state. The second antibody targets the subunit interface involved in binding benzodiazepines and antagonizes diazepam potentiation. We identify key residues in these antibodies involved in specificity and affinity and confirm structure-based hypotheses for functional effects using electrophysiology. Together these studies define mechanisms of direct functional antagonism of neurotransmission underlying autoimmune encephalitis in a human patient.

Keywords: Autoimmune disease, cryo-electron microscopy, GABAA receptor, Cys-loop receptor, encephalitis

Graphical Abstract

graphic file with name nihms-1818220-f0001.jpg

In Brief:

Structural analysis of antibodies from an encephalitis patient bound to the GABAA receptor reveals mechanisms of channel inhibition and pathology that drive autoimmune encephalitis in humans.

Introduction

Autoimmunity arises when the body’s T-cells or antibodies attack self-antigens. The trigger for this improper immune response can be environmental, such as a virus, or internal, such as a tumor. The consequences of autoimmune disease can be short-lived and self-resolving, or can become chronic, debilitating, and lead to death. Autoantibodies targeting a variety of tissues have been investigated for decades (1988; Obermayer-Straub et al., 2000). However, the study of neuropsychological disorders directly mediated by anti-neuronal antibodies is in its infancy. The first case of anti-NMDA receptor encephalitis was described in 2007 (Dalmau et al., 2007), and represents one entity in the rapidly growing disease category of autoimmune encephalitis (Prüss, 2021). Autoimmune encephalitis is characterized by severe and acute-onset symptoms comprising psychosis, refractory seizures, movement disorders, impaired consciousness and dysautonomia. Established antibodies in serum or cerebrospinal fluid from affected patients can now be routinely detected in cell-based assays overexpressing the respective proteins. Novel clinically relevant antibodies are continuously discovered, and the antigenic targets identified using immunoprecipitation and mass spectrometry. Anti-neuronal targets associated with pathological disease include ionotropic and metabotropic glutamate receptors, Leucine Glioma Inactivated 1 (LGI1), contactin-associated protein-like 2 (CASPR-2), dopamine receptors, and metabotropic and ionotropic γ-amino butyric acid (GABA) receptors (GABAB and GABAA, respectively) (Dalmau and Graus, 2018; Prüss, 2021).

GABAA receptors are part of the greater Cys-loop receptor family of pentameric ligand-gated ion channels. The GABAA receptors form anion-selective channels, and generally act to dampen neuronal excitability. They are usually heteromeric, assembling from a panel of 19 subunits. The most common isoform at synapses comprises two α1 subunits, two β subunits, and one γ2 subunit (Nutt and Malizia, 2001) (Figure 1). Each subunit is positioned symmetrically or pseudo-symmetrically around the axis of the ion pore. The neurotransmitter GABA binds in the extracellular domain (ECD) at β-α subunit interfaces. This binding triggers conformational changes that open an ion channel formed by the second helix (M2) of the transmembrane domain (TMD). In the sustained presence of agonist, the channel adopts a non-conducting desensitized state (Gielen and Corringer, 2018). When GABA concentrations decrease, GABA dissociates and the channel returns to the resting, activatable state. Inhibiting this inhibitory receptor leads to nervous system hyperexcitability, which can result in anxiety, seizures, catatonia, and death. These ends may be achieved through receptor internalization, or through stabilizing one of the closed-channel states.

Figure 1: Encephalitis-associated autoantibodies bound to the synaptic GABAA receptor.

Figure 1:

A, Side and top views of the Fab115: GABAA complex cryo-EM map. Subunits and Fab densities are indicated. The constant region of the Fab is not shown as this peripheral domain is flexible and conformationally disordered. B, Same as for A, but with the Fab175.

As with other autoimmune encephalitides, target identification of the GABAA receptor is the essential starting point for understanding, diagnosing and treating disease (Petit-Pedrol et al., 2014). However, it reveals little about how the improper immune response causes pathology. For different forms of autoimmune encephalitis, the mechanism of the autoantibodies’ pathogenicity is mainly attributed to target receptor clustering and internalization (Dalmau et al., 2017). Direct autoantibody effects on neuronal function have only recently been appreciated (Crisp et al., 2019). Isolation of monoclonal antibodies from the CSF of an 8-year-old encephalitis patient identified at least two different autoantibodies that cause seizures and status epilepticus in the absence of receptor internalization (Kreye et al., 2021). Here, we use structural biology to reveal the pathogenic mechanisms of these patient-derived anti-neuronal antibodies bound to their physiological target, the GABAA receptor.

Results

Identification of stable autoantibody-GABAA receptor complexes

We tested a panel of five patient-derived antibodies (Kreye et al., 2021) for binding to the purified α1β2γ2 GABAA receptor (Figure S1, refers to Figure 1). Purified monoclonal antibodies were first screened for receptor binding by fluorescence-detection size exclusion chromatography (FSEC) (Kawate and Gouaux, 2006). This assay detected binding by all antibodies tested except for mAb198 (Figure S1A). Fab fragments were then generated from antibodies mAb101, mAb115, mAb175 and mAb201 for structural studies. Binding stoichiometries were estimated by FSEC with increasing molar ratios of Fab to receptor (Figure S1BE, refers to Figure 1) and reactivity to native receptors was analyzed in murine brain tissues (Figure S1F, refers to Figure 1) before large-scale complex formation and purification. Fab201 failed to bind detectably to the GABAA receptor and was excluded from structural studies. Fab101 no longer bound the GABAA receptor with an affinity suitable for structural work; it may require bivalency to bind appreciably. We thus pursued both Fab115 and Fab175 in structural studies.

Structure of the GABAA receptor bound to an inhibitory autoantibody

In contrast to mechanisms of autoantibody-induced neurological disease described thus far (Hughes et al., 2010; Kreye et al., 2016; Petit-Pedrol et al., 2014), mAb115 and Fab115 directly inhibit GABAA receptor function without causing receptor internalization from the cell surface (Kreye et al., 2021). We were intrigued by this phenomenon and sought to understand its mechanism of inhibition by obtaining the structure of Fab115 bound to the GABAA receptor. We purified the α1β2γ2 synaptic GABAA receptor and reconstituted it into brain lipid nanodiscs (Kim et al., 2020). Reconstituted receptors were incubated with Fab115 before further polishing by size-exclusion chromatography (Figure S1G,H). We applied the purified complex to cryo-electron microscopy (cryo-EM) grids and collected a dataset that allowed us to reconstruct a 3.0 Å resolution map (Figures 1A and S2, Table S1). The map quality allowed us to confidently model the receptor and the variable domains of the Fab (VH and VL for heavy and light chain variable domains, respectively). Local resolution in the areas of greatest interest, including side-chain interactions between Fab115 and receptor, was ~2.9 Å (Figure S2I, S3 and Video S1, refer to Figures 1, 2, 3).

Figure 2: Fab115 competes directly with neurotransmitter binding.

Figure 2:

A,B Side and top views of receptor and Fab (gold) atomic models. Boxes indicate expanded regions shown in panels C and D. C, Fab-receptor interactions in the neurotransmitter binding site. Key amino acids are highlighted; dashed lines indicate putative electrostatic interactions closer than 4 Å. D, Anchoring interaction between Fab115 and GABAA. Residues shown are an alignment of key interacting residues of α subunits; red letters indicate potential specificity-determining residues. E, Subunit interface contacts with Fab. One β2:α1 subunit interface is shown; red box indicates neurotransmitter binding site and spheres indicate amino acids contacted by Fab115. Inset table shows surface area buried by Fab heavy and light chains on respective GABAA receptor subunits.

Figure 3: Fab175 binds the GABAA receptor α-γ interface.

Figure 3:

A, Overview of model with area shown in panels B and C indicated. B, Amino acid interactions between Fab175 and the α1-γ2 interface of the GABAA receptor. C, The interactions between the lower part of the Fab and the α1 benzodiazepine binding pocket Loop C. D, Subunit interface contacts with Fab. One β2:α1 subunit interface is shown; red box indicates neurotransmitter binding site and spheres indicate amino acids contacted by Fab175. Inset table shows surface area buried by Fab heavy and light chains on respective GABAA receptor subunits.

The complex structure reveals one Fab115 bound at each of the two β-α subunit interfaces in the ECD (Figure 1A, Figure 2, Figures S3,S4). The complementarity determining region (CDR) 3 of the Fab heavy chain penetrates the neurotransmitter binding pocket and contacts conserved residues known to impact agonist binding (Figure 2B, Figures S35, refer to Figures 2, 3). In particular, R105 of the VH CDR3 is positioned to make electrostatic and cation-π interactions with several residues on the β2 and α1 subunits (Figure 2C). Of special interest is the contact between VH R105 and β2 E155. E155 forms a salt bridge with the primary amine of the neurotransmitter GABA and is critical for binding and gating (Jatczak-Sliwa et al., 2020; Mortensen et al., 2014; Newell et al., 2004). Other residues lining the neurotransmitter binding site that interact with CDR3 of Fab115 have also been implicated in binding to GABA, including the following on the β subunit: Y97 (Boileau et al., 2002), Y157 (Amin and Weiss, 1993; Newell and Czajkowski, 2003; Newell et al., 2004), S156 (Newell et al., 2004), F200 (Zhu et al., 2018), and Y205 (Amin and Weiss, 1993; Zhu et al., 2018). Thus, the Fab115 VH CDR3 mimics an aspect of neurotransmitter chemistry and likely competes with GABA binding in a manner analogous to a competitive antagonist (Kim et al., 2020; Masiulis et al., 2019).

In the initial antibody characterization, mAb115 was found to be selective for binding among different GABAA receptor subunits (Kreye et al., 2021). The structure reveals the basis of this exquisite selectivity. First, although GABAA β subunits can form a homopentamer, β alone did not bind antibody despite its numerous contacts with Fab115 in the neurotransmitter binding pocket (Kreye et al., 2021). Examination of the Fab115:receptor structure reveals that most of the interactions between Fab115 and receptor are with the α1 subunit (Figures 2E, S5); contacts with β alone cannot support binding. Furthermore, mAb115 is only able to bind GABAA receptors containing the α1, but not α2–5 subunits. This specificity may arise from interactions between Fab115 and four key residues on the α1 subunit: E170, R173, E174, and R177 (Figure 2D). While only R177 has been studied for functional importance (Newell and Czajkowski, 2003), evidence for the contribution of all these residues to antibody binding specificity is found in an examination of contacts between Fab115 and the receptor (Figure S5, yellow highlights). Most contacted residues are conserved among α1-α6; as mAb115 can only bind α1, residues shared across subunits would not be expected to be sufficient for antibody binding. The exceptions are T122-D124 and E170-V180. In the case of T122-D124, Fab115 only makes electrostatic interactions with backbone carbonyl oxygens, and therefore the identity of the residues present in the receptor are less important. The string of 11 residues beginning at E170 exhibits variable conservation (Figure 2D); however, the tetrad of E170, R173, E174 and R177 is only found in α1, and all four residues are involved in salt bridges. These electrostatic contacts are poised to stabilize the Fab-receptor interaction and likely determine the selectivity of Fab115 for α1-containing receptors.

Structural and sequence analyses suggest the ways in which mAb115 evolved to increase not just its selectivity but also its affinity for the GABAA receptor. Somatic hypermutation is a process unique to B cells and antibody secreting cells that diversifies immunoglobulins in their variable gene regions as an adaptive immune response to enhance affinity for their antigenic target. We compared the Fab115 antibody sequence to an immunoglobulin gene database to derive a germline ancestor with highest sequence identity. This approach identifies somatically hypermutated residues within the variable V(D)J gene segments that have evolved for affinity maturation (Figure S6, (Reincke et al., 2020)). Both Fab115 VH and VL contain somatically hypermutated residues that are involved in binding the GABAA receptor (Figure S6, refers to Figures 13, S3). These include R97 and K98 of the Fab115 VL, which are encoded in the germline sequence as an asparagine and a glycine. These mutations contribute two new positive charges at each interface that are stabilized at the interface through salt bridges to help anchor Fab115 to the α1 subunit (Figure 2D). Furthermore, D52 of the Fab115 VH is an asparagine in the germline antibody sequence (Figures 2D, S6). This mutation from a polar residue to a charged residue would generate a stronger molecular interaction, changing it from a hydrogen bond to a salt bridge. Thus, somatic hypermutations are predicted to enhance the affinity of Fab115 for its target.

Structure of the GABAA receptor in complex with a non-competitive autoantibody

The mAb175 autoantibody also triggered seizure activity in rodents and was shown to require both the α1 subunit and the γ2 subunit for binding to the GABAA receptor in cell-based assays (Kreye et al., 2021). The α-γ interface is of special interest because it forms the binding site for benzodiazepines, a modulatory class of GABAA drugs that includes anxiolytics and sedative-hypnotics (Sigel and Ernst, 2018), and is an important site for endogenous modulators, termed endozepines (Tonon et al., 2020). In accordance with requirement of the α-γ interface for mAb175 binding, FSEC experiments indicated a stoichiometry of 1:1 for receptor:Fab175 (Figure S1D, refers to Figures 3,S2,S3). We used cryo-EM to obtain a structure of the Fab175:GABAA complex at an overall resolution of 3.0 Å (Figure 1B, Figure S2H, Table 1). The map quality allowed us to confidently model the receptor and the variable domains of the Fab (Figure S3GJ).

Fab175 binds at the α1-γ2 subunit interface, consistent with cell-based and FSEC experiments (Figures 1B, 3A, S3GJ, S4CD). In a manner similar to Fab115, the majority of surface area contacted by Fab175 is with the α1 subunit (Figures 3D, S5). Fab175 VH contains a methionine (M103) that buries itself into a hydrophobic pocket composed of residues from both sides of the α1:γ2 interface, above the benzodiazepine site (Figures 3B, S3H,J). The Fab175 VL docks onto Loop C of the α1 subunit; this loop caps the benzodiazepine-binding pocket of the receptor (Figures 3C, S3G). This VL attachment site centers upon α1 R164 of the GABAA receptor, which forms a network of five electrostatic interactions connecting the two proteins (Figure 3C). This arginine is unique to α1; while some of the other α subunits contain a lysine at this position, a primary amine would not recapitulate the elaborate network of electrostatic interactions. This arginine likely contributes to subunit specificity, as most other receptor residues contacted by Fab175 are conserved (Figure S5, refers to Figures 13)). Fab175 underwent somatic hypermutation to a lesser degree than Fab115, changing six amino acids in the heavy chain and one in the light chain (Figure S6, refers to Figures 13)).

Autoimmune encephalitis Fabs act as competitive and allosteric antagonists

The conformational states observed in the receptor structures shed light on how autoantibody binding triggers disease pathology. We first compared the two autoantibody complexes with the receptor bound to GABA alone (Figure 4A, PDB 6X3Z, (Kim et al., 2020)). Fab115, through direct interactions, stabilizes the neurotransmitter site Loop C in a widely open conformation at β-α interfaces akin to its conformation in the presence of competitive antagonists (Figure 4B). Unlike the Fab115-bound receptor, the Fab175-bound receptor contains density consistent with GABA in its binding site, even though the agonist was not added to the preparation. The presence of GABA induces a partial closure of Loop C in the neurotransmitter binding pockets (Figure 4B). Normally, closure of Loop C by agonist triggers a rotation and contraction of the ECD, which is then translated into opening of the transmembrane pore (Kim and Hibbs, 2021; Nemecz et al., 2016). After channel opening, the agonist remains bound but a lower gate in the pore closes, a process termed desensitization. Indeed, in a similar preparation, the presence of endogenous GABA caused the transmembrane domain to adopt a desensitized-like conformation (Laverty et al., 2019). However, the Fab175-bound GABAA structure reveals an interruption in this state transition (Figure 4E, F). Although Loop C is mostly closed, the ECD of the Fab175 complex is not contracted. Fab175 blocks this contraction by wedging between the α1 and γ2 subunits in the upper part of the ECD (Figure 3B). Superposition of the Fab175 bound structure on the GABA-bound structure suggests many atomic clashes would prevent the ECD contraction that initiates channel gating in the pentameric receptor superfamily (Figure 4G, (Lefebvre et al., 2021). The consequence is that GABA is positioned farther from the residues of the neurotransmitter binding pocket. Specifically, the GABA amino nitrogen is 4.1–4.5 Å from E155, as opposed to 2.7–3.1 Å in the GABA-bound structure in the absence of Fab (range covers the two GABA sites). The GABA-E155 interaction is required for converting agonist binding into channel gating (Newell et al., 2004), and is weakened in the presence of Fab175. Together, the consequences of both Fab115 and Fab175 binding are stabilization of the receptor pore in a resting-like, non-conducting conformation, with the classical activation gate at the 9ʹ position closed (Figure 4D, F, H). The structural findings thus suggest a direct competition mechanism for Fab115 and an allosteric antagonism mechanism for Fab175, with both stabilizing a resting-like state of the ion channel.

Figure 4: Conformational Differences between Fab115-bound, Fab175-bound, and GABA-bound receptors.

Figure 4:

A, Side view of Fab115-bound (bright green, dark blue, magenta), Fab175-bound (light green, light blue, pink), and GABA-bound (gray, PDB 6X3Z (Kim et al., 2020)). Boxes indicate areas represented in indicated panels. B, Detail of Loop C displacement at a β2-α1 interface; mesh indicates residual density for GABA. C, Top view of the ECD comparing Fab115-bound vs Fab175-bound. Arrows indicate binding sites of Fab175 (red) and Fab115 (gold). D, Same as in C, but comparing cylinder representations of the TMD. E, Top view of the ECD comparing Fab175-bound vs GABA-bound. F, same as in E but comparing cylinder representations of the TMD. G, Fab175 docked into GABA-bound receptor structure (6X3Z); spheres indicate side chains that clash. H, Pore radius analysis for a panel of GABAA receptor structures. GABAA + Fab175 and GABAA + Fab115, this study. 6I53 and 6X3Z are agonist-bound (Kim et al., 2020; Laverty et al., 2019). 6HUJ and 6X3S are antagonist-bound (Kim et al., 2020; Masiulis et al., 2019).

We sought to probe the structure-based autoantibody mechanisms using functional measurements. Initial electrophysiological studies on autaptic neurons found that mAb115 but not mAb175 directly inhibited GABA-activated currents (Kreye et al., 2021). We performed whole cell patch clamp experiments on transfected cells expressing the α1β2γ2 receptor to test the concentration dependence of Fab inhibition. These experiments revealed, first, that Fab115 binds effectively irreversibly (Figure 5A). Application of GABA after exposing cells to increasing concentrations of Fab revealed a clear dose-dependence in inhibition (Figure 5B). Tests with Fab175 revealed no substantial inhibition of GABA activation, consistent with the earlier results testing inhibition by mAb175 on autaptic neurons. Nonetheless, when the antibody was injected into mouse CNS, it triggered spontaneous epileptic events (Kreye et al., 2021). The structural information suggested that Fab175, through binding at the α-γ interface near the benzodiazepine site, could affect the activity of modulators acting there. We thus tested the effect of Fab175 on diazepam potentiation (Figure 6). We found that Fab175, in a concentration dependent manner, inhibits diazepam potentiation of GABA activation (Figure 6B). A speculative mechanism for Fab175 antagonism of receptor activity may thus involve altering the way endogenous modulators (endozepines) act through this site, with effects on GABA activity too subtle to detect in our patch clamp experiment. One prominent endozepine is the diazepam binding inhibitor (DBI), which is a benzodiazepine site ligand that was recently found to be an anti-epileptic positive modulator in the thalamic reticular nucleus (Christian et al., 2013). Antagonism of this interaction, as well as other positive modulator endozepines (Tonon et al., 2020), may be a component of how this antibody triggers disease pathology; studies with GABAA subunit knockout mice could further substantiate this hypothesis. Taken together, these findings reveal distinct mechanisms leveraged by the two antibodies to cause autoimmune encephalitis with seizures.

Figure 5: Fab115 irreversibly antagonizes GABAA receptor activity.

Figure 5:

A, Representative whole cell patch-clamp electrophysiology recordings from transfected HEK cells. Response to GABA activation is shown before and after perfusion of Fab115. B, Bar graph showing mean responses with standard deviation for the GABA response in the absence (control) and presence of increasing concentrations of Fab115. T-test **** p<0.0001, *** p<0.001. Biological replicate numbers (individual cells) are shown in the graph. C, Cartoon model representing a proposed mechanism of GABAA receptor antagonism by Fab115.

Figure 6: Fab175 inhibits benzodiazepine-mediated potentiation of GABAA receptors.

Figure 6:

A, Representative whole cell patch-clamp electrophysiology recordings from transfected HEK cells. Response to GABA activation in the absence and presence of diazepam is shown, followed by the response to GABA plus diazepam following application of Fab175. B, Bar graph showing mean responses with standard deviation for the fractional diazepam potentiation before (control) and after application of Fab175. T-test * p=0.64, not significant; *** p=0.0006. Biological replicate numbers (individual cells) are shown in the graph. C, Cartoon model representing a proposed mechanism of action of Fab175 at the GABAA receptor.

Discussion

The extent to which humoral autoimmunity in the CNS triggers disease has only recently begun to be appreciated. To date there have been no structural studies on how disease-causing autoantibodies from patients act on their neurotransmitter receptor targets. Here we determined structures of the GABAA receptor bound to two patient-derived pathogenic antibody fragments. The structures reveal how the antibodies bind at distinct subunit interfaces to interfere with receptor function. Mapping of the interactions identified residues important for the specificity of the human antibodies for restricted subtypes of GABAA receptors. We defined conformational changes that are stabilized or inhibited in the presence of autoantibodies by comparison to established GABAA receptor structures. These structural analyses combined with electrophysiological measurements provide mechanisms for direct and allosteric antagonism of inhibitory neurotransmission by autoimmune antibodies and can explain the clinical symptoms of patients with GABAA receptor encephalitis at the atomic level.

Limitations of the Study

In this study we determined two structures of receptor-bound recombinant antibodies that were isolated from an individual patient in the acute phase of an autoimmune encephalitis. While we have shown functional effects of both Fabs that correlate with structural predictions, systematic mutagenesis would be important to confirm the relative importance of individual determinants of antibody-receptor binding. Further, we cannot draw broad conclusions about effects from other GABAA receptor autoantibodies from this individual and other patients. In a previous study, these antibodies recapitulated a severe clinical phenotype reminiscent of autoimmune encephalitis with the predominant symptom of seizures in rodent models without causing receptor internalization. It is possible that other GABAA receptor autoantibodies in this patient and in separate autoimmune encephalitis cases work through receptor cross-linking and internalization. Our findings support that cross-linking and internalization are not the only mechanisms of autoimmune antibodies targeting neuronal receptors and draw attention to a spectrum of alternate mechanisms contributing to autoimmune pathology.

STAR Methods

Resource Availability

Lead Contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Ryan E. Hibbs (ryan.hibbs@utsouthwestern.edu).

Materials Availability

All unique reagents generated in this study are available from the Lead Contact with a completed Materials Transfer Agreement.

Data and Code Availability

  • Cryo-EM maps and atomic model coordinates have been deposited in the EMDB and RCSB, respectively, under the accession codes 7T0W/EMD-25583 and 7T0Z/EMD-25585. They will be publicly available as of the date of publication.

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

Experimental Model and Subject Details

Cell Lines

The Sf9 insect cells were obtained from the ATCC (Cat# CRL-1711) and cultured in an InnovaR shaking incubator at 27°C at 130 rpm with Sf900 III media (Thermo Fisher Scientific). HEK293S GnTI-suspension cells were obtained from the ATCC (Cat# CRL-3022) and cultured in a Thermo Scientific Reach-In CO2 Incubator at 37°C with 8% CO2 at 130 rpm. All cell lines used in this study are female in origin.

Method Details

Fab production and purification

Fab fragments were produced as previously described (Kreye et al., 2021). cDNA encoding the heavy and light chains of the immunoglobulins was isolated via reverse transcription and specific amplification from single cerebrospinal fluid cells sampled from a pediatric patient in the acute phase of GABAA receptor encephalitis. These sequences were cloned into mammalian expression vectors, from which monoclonal antibodies were recombinantly expressed in HEK293T cells and purified from cell culture supernatant for initial characterization. Fab fragments were generated by replacing the Fc domains CH2 and CH3 of the heavy chain expression vector with a FLAG-Tag and a His-Tag following the amino acids PKSCDKTH of the hinge region. Fab fragments were expressed, purified using affinity chromatography via the His-Tag and controlled for purity using SDS Page by an external provider (inVivo BioTech, Germany).

Immunohistochemistry

Immunohistochemical stainings of murine brain tissue were performed as previously described (Kreye et al., 2016; Kreye et al., 2021). In brief, 20 μm sagittal cryosections were fixed with paraformaldehyde (PFA), first blocked in PBS supplemented with 2% BSA (Dianova) and 5% Normal Goat Serum (Abcam) at room temperature for one hour, then incubated with 20 μg/ml of Fab at 4°C overnight, then with an anti-human Fab-specific secondary antibody (Dianova, 1:500) at room temperature for 2 hours, and finally with DRAQ5 (Thermo Fisher Scientific, 1:500) at room temperature for 10 minutes. Confocal images were recorded at an inverted fluorescence microscope (Leica DMI8) with identical conditions for all Fabs.

Receptor expression, purification, and reconstitution into nanodiscs

To express the GABAA receptor, we generated bacmam virus from a tri-cistronic construct encoding the α1, β2 and γ2 subunits of the GABAA receptor (Kim et al., 2020). These subunits contained a deletion in the intracellular domain between transmembrane helices M3 and M4. These amino acids were replaced with an SQPARAA linker; this deletion does not affect functional behaviors in electrophysiology and functional assays (Kim et al., 2020; Zhu et al., 2018). Bac-mam viruses were made in Sf9 cells (Vaughn et al., 1977), and the tric-cistronic virus was transduced at an MOI of 1 into HEK293S GnTI suspension cells (Reeves et al., 2002) at a density of 3–4 × 106/ml. The cells were cultured at 30 °C for 72 h before harvesting by centrifugation. Cells were washed once in 20 mM Tris, pH 7.4, 150 mM sodium chloride (TBS) and recentrifuged. After resuspension in TBS + 1 mM phenylmethanesulfonyl fluoride (PMSF; Sigma), cells were lysed by passage through an Avestin Emulsiflex at 5,000–15,000 psi. Lysed cells were first centrifuged at low speed (10,000 g) to remove nuclei and unlysed cells, then centrifuged at 186,000 g for 2 h to pellet membranes. Membranes were stored at −80 °C until used. To purify proteins, approximately 5 g of membranes were Dounce homogenized in TBS + 1 mM PMSF. The detergent n-dodecyl-β-D-maltoside (DDM; Anatrace) was added to a final concentration of 40 mM, and solubilization was performed at 4 °C while nutating. Insoluble material was removed by centrifugation at 186,000 g for 40 min. The supernatant containing solubilized protein and its native lipids was bound to Strep-Tactin Superflow high-capacity resin (IBA Life Sciences) via gravity flow. The resin was washed with TBS plus 1 mM DDM, supplemented with 1% Porcine Brain Polar Lipids (Avanti) to promote receptor stability. The GABAA receptor protein was eluted in the same wash buffer supplemented with 5 mM desthiobiotin (Sigma).

Purifications of the GABAA receptor were done in the absence of exogenous ligand. For autoantibody FSEC experiments shown in Supplemental Figure 1A, purified receptor reconstituted in lipidic nanodiscs was aliquotted and flash frozen in liquid nitrogen until required. A GABAA receptor aliquot was thawed on ice, and ~2 μg of receptor was incubated with ~0.5–2 μg of each Fab or mAb at room temperature for 10 min. Protein was diluted in TBS (20 mM Tris, pH 7.4, 150 mM NaCl) and centrifuged to pellet aggregated receptor before analysis by FSEC. The Fab stoichiometry screening shown in Figure S1BE was performed in a similar manner, with increasing molar ratios of Fab to receptor.

Cryo-EM Sample Preparation

For Cryo-EM, the GABAA receptor protein reconstituted in nanodiscs was concentrated to ~500 μl. The concentrated protein was divided into 50μl aliquots and preincubated with porcine brain polar lipids for 10 minutes at room temperature. Saposin (Lyons et al., 2017) was then added for two minutes at room temperature. Porcine brain polar lipids and saposin were added to the purified protein at a molar ratio of 1 GABAA protein: 30 saposin protein: 230 lipid. After the addition of the saposin the mixture was diluted to 500 μl for each aliquot with TBS to initiate reconstitution. This mixture nutated at 4°C overnight in the presence of bio-beads (Bio-Rad) to remove detergent at a concentration of 100mg/ml. The removal of detergent was continued the next morning with a fresh batch of bio-beads at 4 °C for one hour, after which incorporation was assayed by fluorescence-detection size-exclusion chromatography (Kawate and Gouaux, 2006). Fab was added to the receptor at molar ratios of 1:3 (for Fab115) or 1:2 (for Fab175). The complex was further polished by size exclusion chromatography using TBS as a mobile phase. The best fractions, as assessed by FSEC, were pooled and concentrated to ~4–6 mg/ml before freezing for cryo-electron microscopy (cryo-EM).

For preparation of cryo-EM grids, 200-mesh Cu 1.2/1.3 grids from Quantifoil were glow-discharged in a Pella glow discharge apparatus for 30 mA/80s on top of a metal grid holder (Ted Pella). To induce random particle orientations, protein was supplemented with 0.5 mM fluorinated fos-choline-8 (Anatrace) immediately before freezing. Grids were frozen with a Mark IV Vitrobot (FEI/ThermoFisher). Three μl of protein were applied to grids before blotting for 3.5 s at 100% humidity and 4 °C. Frozen grids were stored in liquid nitrogen until used.

Cryo-EM data collection and processing

The electron microscopy was performed on a 300kV Titan Krios (FEI/Thermo-Fisher) equipped with a K3 direct electron detector and a GIF energy filter (20 eV) using super-resolution mode (Gatan) and collected via SerialEM (Mastronarde, 2005). The approximate physical pixel size was 1.079 Å/pixel. The total dose was 36–86 e2.

All data were processed within the Relion 3.0.8 suite of software (Zivanov et al., 2018). Dose-fractionated images were collected using SerialEM (Mastronarde, 2005). Data processing is described in Supplemental Figure 3. In brief, micrographs were motion corrected and gain normalized using MotionCor2 (Zheng et al., 2017). Contrast transfer function (CTF) and defocus values were estimated using GCTF (Zhang, 2016). Particles were picked using CrYOLO (Wagner et al., 2019). Following 2D and 3D classification, particle orientations were refined and subjected to Bayesian polishing, all performed in Relion (Zivanov et al., 2018). Focused 3D classification was performed using the refined angles on polished particles. For the Fab175 dataset, we performed a further 3D classification focused on the γ-TMD, which is more mobile in some purified sample preparations (Zhu et al., 2018). The Fab115 γ-TMD classes had equivalent density, and this extra step was unnecessary. Programs were compiled by SBGrid (Morin et al., 2013).

Model building and refinement

Statistics for each dataset are shown in Supplemental Table 1. The bicuculline-bound α1β2γ2 structure (PDB:6X3S) was used as a starting model for the GABAA receptor. To generate starting models for the Fabs used in this study, amino acid sequences were submitted to the SwissModel (Waterhouse et al., 2018) and PIGS (Waterhouse et al., 2018) servers. The top hits for Fab175 and Fab115 were modeled on PDB IDs 4HH9 and 5WL2.1 (Bryson et al., 2016), respectively. These homology models were manually docked and then fitted into maps using UCSF Chimera (Pettersen et al., 2004). Real-space refinement was performed via iterations of local and global adjustment in Coot (Emsley et al., 2010) and Phenix (Liebschner et al., 2019) respectively. Model quality was assessed in MolProbity (Williams et al., 2018). For placement of important side chains such as E174 and K98 (of the receptor α1 and Fab115 Light Chains, respectively), cryo-EM maps were high-pass filtered to 6.0 Å using Relion Image Handler (Gharpure et al., 2019). The pore profiles shown in Figure 4 were created using HOLE (Smart et al., 1996); hydrophobicity plots were created with CHAP (Klesse et al., 2019).

Analysis of Antibody Sequences to Germline

Variable antibody sequences were compared using custom BASE software (Reincke et al., 2020) to human immunoglobulin germline V(D)J gene segments to identify genes with highest sequence identity.

Electrophysiology

Whole cell voltage-clamp recordings were made from adherent HEK293S GnTI cells (ATCC CRL3022) transiently transfected with the tri-cistronic pEZT construct used for structural analysis. Each well of cells in a 12-well dish was transfected with 0.1–0.3 μg plasmid using Lipofectamine 2000 Reagent (Invitrogen). The transfected cells were incubated at 30°C. 24 h post-transfection, cells were re-plated on 35 mm dishes and allowed to settle for at least 2 hours. Recordings were made 24–96 hours after transfection. Bath solution contained (in mM): 140 NaCl, 2.4 KCl, 4 MgCl2, 4 CaCl2, 5 HEPES and 10 glucose pH 7.3. Borosilicate pipettes were pulled and polished to an initial resistance of 2–4 MΩ and filled with the pipette solution containing (in mM): 100 CsCl, 30 CsF, 10 NaCl, 10 EGTA, and 20 HEPES pH 7.3. The recordings were made with an Axopatch 200B amplifier, sampled at 5 kHz, and low-pass filtered at 2 kHz using a Digidata 1440A (Molecular Devices) and analyzed with pClamp 10 software (Molecular Devices). Cells were held at −75 mV. Solution exchange was achieved using a gravity driven RSC-200 rapid solution changer (Bio-Logic). For short application (60–80 s pre-application), Fab solutions were prepared in bath solution from a concentrated stock solution.

Quantification and Statistical Analysis

Peak currents in whole cell patch clamp electrophysiology experiments were measured in the absence or presence of antibody fragments. Statistical analysis was performed using GraphPad Prism 9.2.0 software (GraphPad software, Inc, La Jolla, CA). Data are expressed as means ± S.D, and Welch’s t-test was used. A p-value of ≤ 0.05 was considered statistically significant (**** p ≤ 0.0001; ***, p ≤ 0.001; *, 0.01 ≤ p ≤ 0.05) (Figure 5 and 6). Replicate numbers n = 7–9 for Figure 5 and n = 4–12 for Figure 6, from independent cells.

Supplementary Material

1

Supplemental Figure 1: Patient-derived antibodies bind to the α1β2γ2 GABAA receptor and representative purification, related to Figure 1. A, Purified GABAA receptor was reconstituted in nanodiscs in the absence or presence of each monoclonal antibody (mAb) indicated and analyzed by fluorescence-detection size exclusion chromatography (FSEC). A shift to the left indicates an increase in size associated with binding. B-E are screens of selected Fab fragments produced from the mAbs. Increasing amounts of Fab were added to GABAA receptor in nanodiscs to determine at which point the elution volume shift is saturated. F, Immunofluorescence staining of selected GABAA Fabs (green) on PFA-fixed murine brain tissue sections shown in the CA3 region of the hippocampus with DRAQ5 stained nuclei (red). Representative scale bar indicates 50 μm. G, FSEC traces of receptor alone (black trace) and receptor plus Fab115 (blue trace). H, FSEC trace of the final material applied to EM grids; inset shows Coomassie-stained SDS-PAGE gel of the complex.

2

Supplemental Figure 2: Cryo-EM processing workflow for Fab175 + GABAA, related to Figures 1, 3. Each panel represents a different stage of the data collection and processing. A, Representative micrograph. Scale bar indicates 100 nm. B, 2D classification with selected classes boxed in red. C, First 3D classification with selected class indicated. D, First refinement. E, Second 3D classification using the refined angles from D to perform fine local angular searches. F, Second refinement. G, 3D classification on signal subtracted particles to focus alignment on the γ-TMD. H, Final refinement, sharpening, local resolution estimation and FSC Curve for the Fab175+GABAA dataset. I, as in H but for the Fab115+GABAA dataset.

3

Supplemental Figure 3: Quality of Cryo-EM maps at regions of interaction between Fab and receptor, related to Figures 2, 3. A, Overview of Fab115:GABAA complex map from view parallel to membrane. Panel D inset is indicated. B, As in panel A, but turned 180° to show other Fab115:GABAA interface. Inset indicated is shown in panel E. Mesh in panels D and E is the result of high-pass filtering the density maps to 6 Å in order to emphasize higher resolution features. C, Overview of Fab115:GABAA complex map from view of outside the cell looking in. Panel F inset is indicated, highlighting the interaction between the Fab CDR3 and neurotransmitter binding domain. G, Overview of Fab175:GABAA complex map from view parallel to membrane. Panel I inset is indicated. H, Overview of Fab175:GABAA complex map from view of outside the cell looking in. Panel J inset is indicated, highlighting CDR3 of the antibody heavy chain, which inserts into the ECD α1:γ2 interface.

4

Supplemental Figure 4: Overview of receptor:Fab models, related to Figures 14. A, Fab115: GABAA model. ECD, extracellular domain. TMD, transmembrane domain. B, Fab115: HC, heavy chain; LC, light chain. CDR, complementarity determining region. The CDRs are colored in red-orange for contrast. C, D Same as for A, B but CDRs are colored green. E, F Subunit interface contacts with Fab. E, One β2:α1 subunit interface is shown; red box indicates neurotransmitter binding site and spheres indicate amino acids contacted by Fab115. Inset table shows surface area buried by Fab heavy and light chains on respective GABAA subunits. F, Same as for E but for Fab175.

5

Supplemental Figure 5: Alignment of GABAA receptor subunits contacted by autoantibodies, related to Figures 23. Amino acid sequence alignments of GABAA subunits contacted by Fab115 and Fab175 are shown. On the right-hand side of the figure are cartoons indicating the location of the antibodies bound (yellow is Fab115, red is Fab175). Subunits shown in the alignment for each box are colored.

6

Supplemental Figure 6: Somatic hypermutations of Fab115 and Fab175 compared to receptor contacts, related to Figures 23. A, Alignments between sequences of Fab115 and its presumed germline ancestor. Only the variable domain is shown. CDR1, CDR2, and CDR3 are indicated in bold for each antibody segment. Somatic hypermutations are indicated by noting the parental sequence in the germline. Residues of the V(D)J junctions are underlined. Note, these residues cannot reliably be differentiated from germline ancestors. Hence assignment of somatic hypermutations is limited to V and J genes only, and thus not for most part of the highly variable CDR3. Residues of the heavy and light chains that contact the GABAA receptor are indicated and color coded based on subunit. The NG-KR mutations discussed in the text are indicated by asterisks (*). B, Same as in A but for Fab175.

7

Supplemental Table 1: Cryo-EM Data Collection, Processing and Model Statistics, refers to Figures 1, S1, S2.

8

Supplemental Video 1: Overview and zooms of Fab115:GABAA map density, modeling, and quality, refers to Figures 12, S1S3

Download video file (119.5MB, mp4)
9

Supplemental Video 2: Overview and zooms of Fab175:GABAA map density, modeling, and quality, refers to Figures 1, 3, S1S3

Download video file (54.5MB, mp4)

KEY RESOURCES TABLE.

REAGENT or RESOURCE SOURCE IDENTIFIER
Bacterial and Viral Strains
DH5α Thermo Fisher Scientific Cat# C404003
DH10Bac Thermo Fisher Scientific Cat# 10361012
Chemicals, Peptides, and Recombinant Proteins
n-Dodecyl-β-D-Maltopyranoside (DDM) Anatrace Cat# D310
Cholesterol Sigma-Aldrich Cat# C8667
Porcine brain extract polar lipids Avanti Cat# 141101C
Fos-choline-8, fluorinated Anatrace Cat# F300F
Recombinant Fab101 InVivo BioTech, This Paper This Paper
Recombinant Fab115 InVivo BioTech, (Kreye et al., 2021) (Kreye et al., 2021)
Recombinant Fab175 InVivo BioTech, This Paper This manuscript
Recombinant Fab201 InVivo BioTech, This Paper This manuscript
Recombinant mAb101 (Kreye et al., 2021) (Kreye et al., 2021)
Recombinant mAb115 (Kreye et al., 2021) (Kreye et al., 2021)
Recombinant mAb175 (Kreye et al., 2021) (Kreye et al., 2021)
Recombinant mAb198 (Kreye et al., 2021) (Kreye et al., 2021)
Recombinant mAb201 (Kreye et al., 2021) (Kreye et al., 2021)
d-Desthiobiotin Sigma-Aldrich Cat# D1411
Lipofectamine 2000 Thermo Fisher Scientific Cat# 11668019
Freestyle 293 Expression Medium Thermo Fisher Scientific Cat# 12338-018
Saposin (Lyons et al., 2017) (Lyons et al., 2017)
Sf-900 III SFM Thermo Fisher Scientific Cat# 12658-027
DMEM Medium Corning Cat# 10-013-CV
PBS Corning Cat# 21-040-CV
Fetal Bovine Serum EMD Millipore Cat# TMS-013-B
Bovine Serum Albumin (IgG-free, Protease-free) Dianova Cat# 001-000-161
Normal Goat Serum Abcam Cat# ab138478
Phenylmethanesulfonyl fluoride Sigma Cat# PMSF-RO
Bio-beads SM-2 Adsorbent Media Bio-Rad Cat# 1523920
Murine Brain Tissue (Kreye et al., 2016; Kreye et al., 2021) (Kreye et al., 2016; Kreye et al., 2021)
Paraformaldehyde (PFA) Carl Roth Cat# 0335.1
Anti-human IgG (F(ab’)2-fragment-specific Antibody Dianova Cat# 109-545-097
DRAQ5 Thermo Fisher Scientific Cat# 62254
Critical Commercial Assays
Superose 6 Increase 10/300 GL GE Healthcare Cat# 29091596
Strep-Tactin Superflow high capacity resin IBA Life Sciences Cat# 2-1208-500
Sepax SRT-500 5μM 4.6×300mm Sepax Cat# 215500-4630
Deposited Data
Coordinates of GABAA in complex with GABA (Kim et al., 2020) 6X3Z
Coordinates of GABAA in complex with GABA (Kim et al., 2020) 6X3S
Coordinates of GABAA in complex with Megabody 38 (Laverty et al., 2019) 6I53
Coordinates of GABAA in complex with GABA and picrotoxin (Masiulis et al., 2019) 6HUJ
Coordinates of GABAA in complex with Fab175 This manuscript 7T0Z
Coordinates of GABAA in complex with Fab115 This manuscript 7T0W
Cryo-EM map of GABAA in complex with Fab175 This manuscript EMD-25585
Cryo-EM map of GABAA in complex with Fab115 This manuscript EMD-25583
Experimental Models: Cell Lines
HEK GnTI (Reeves et al., 2002) CVCL_A785
Sf9 (Vaughn et al., 1977) CVCL_0549
Recombinant DNA
pEZT-α1β2γ2 tricistronic GABAA receptor (Kim et al., 2020) (Kim et al., 2020)
Software and Algorithms
Serial EM (Mastronarde, 2005) http://bio3d.colorado.edu/SerialEM/
MotionCor2 (Zheng et al., 2017) https://emcore.ucsf.edu/cryoem-software
GCTF (Zhang, 2016) https://www.mrc-lmb.cam.ac.uk/kzhang/Gctf/
CrYOLO (Wagner et al., 2019) https://cryolo.readthedocs.io/en/stable/
Relion 3.0.8 (Zivanov et al., 2018) https://www3.mrc-lmb.cam.ac.uk/relion/index.php/Main_Page
UCSF Chimera (Pettersen et al., 2004) https://www.cgl.ucsf.edu/chimera/
Phenix (Afonine et al., 2018) https://www.phenix-online.org/
BASE Software (Reincke et al., 2020) https://github.com/automatedSequencing/BASE
Coot (Emsley et al., 2010) https://www2.mrc-lmb.cam.ac.uk/personal/pemsley/coot/
Pymol PyMOL http://www.pymol.org
CHAP (Klesse et al., 2019) https://www.channotation.org/
HOLE (Smart et al., 1996) http://www.holeprogram.org/
pClamp 10 Molecular Devices https://www.moleculardevices.com/products/axon-patch-clamp-system/acquisition-and-analysis-software/pclamp-software-suite
Prism 8 GraphPad https://www.graphpad.com/scientific-software/prism/
Swissmodel (Waterhouse et al., 2018) https://swissmodel.expasy.org/
PIGS (Waterhouse et al., 2018) http://arianna.bio.uniroma1.it/pigs
MolProbity (Williams et al., 2018) http://molprobity.biochem.duke.edu/index.php
Acquire Bruxton https://www.bruxton.com/products.html
TAC 4.2.0 Bruxton https://www.bruxton.com/products.html
Other
Quantifoil Holey Carbon Grids, 1.2/1.3 Cu 200 Mesh Electron Microscopy Sciences Q2100CR1.3

Highlights of “Structural mechanisms of GABAA receptor autoimmune encephalitis”.

  • GABAA receptor structures with pathogenic antibodies from an encephalitis patient.

  • Direct or allosteric receptor inhibition reflect clinical symptoms.

  • Contacts include antibody residues that underwent somatic hypermutation.

  • Electrophysiology supports structure-predicted inhibition mechanisms.

Acknowledgements

We thank R. Cabuco and L. Baxter for baculovirus production and cell culture. We also thank L. Baxter for superb assistance with figures. We thank D. Legesse and J.J. Kim for providing cell membranes. We thank J.J. Kim for assistance in grid freezing. We thank S. van Hoof for assistance with Fab cloning. Single-particle cryo-EM data were collected at the University of Texas Southwestern Medical Center Cryo-Electron Microscopy Facility, which is supported by the CPRIT Core Facility Support Award RP170644. This work was supported by grants from the Welch Foundation (I-1812) and the NIH (DA047325) to R.E.H, and by the German Research Foundation (DFG) (grants FOR3004, PR 1274/3-1, and PR 1274/5-1), by the Helmholtz Association (HIL-A03) and by the German Federal Ministry of Education and Research (Connect-Generate 01GM1908D) to H.P.

Footnotes

Declaration of Interests

The authors declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. (1988). Autoimmune thyroid disease and thyroid antibodies. Lancet 1, 1261–1262. [PubMed] [Google Scholar]
  2. Amin J, and Weiss DS (1993). GABAA receptor needs two homologous domains of the beta-subunit for activation by GABA but not by pentobarbital. Nature 366, 565–569. [DOI] [PubMed] [Google Scholar]
  3. Boileau AJ, Newell JG, and Czajkowski C (2002). GABA(A) receptor beta 2 Tyr97 and Leu99 line the GABA-binding site. Insights into mechanisms of agonist and antagonist actions. J Biol Chem 277, 2931–2937. [DOI] [PubMed] [Google Scholar]
  4. Bryson S, Thomson CA, Risnes LF, Dasgupta S, Smith K, Schrader JW, and Pai EF (2016). Structures of Preferred Human IgV Genes-Based Protective Antibodies Identify How Conserved Residues Contact Diverse Antigens and Assign Source of Specificity to CDR3 Loop Variation. J Immunol 196, 4723–4730. [DOI] [PubMed] [Google Scholar]
  5. Christian CA, Herbert AG, Holt RL, Peng K, Sherwood KD, Pangratz-Fuehrer S, Rudolph U, and Huguenard JR (2013). Endogenous positive allosteric modulation of GABA(A) receptors by diazepam binding inhibitor. Neuron 78, 1063–1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Crisp SJ, Dixon CL, Jacobson L, Chabrol E, Irani SR, Leite MI, Leschziner G, Slaght SJ, Vincent A, and Kullmann DM (2019). Glycine receptor autoantibodies disrupt inhibitory neurotransmission. Brain 142, 3398–3410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Dalmau J, Geis C, and Graus F (2017). Autoantibodies to Synaptic Receptors and Neuronal Cell Surface Proteins in Autoimmune Diseases of the Central Nervous System. Physiol Rev 97, 839–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Dalmau J, and Graus F (2018). Antibody-Mediated Encephalitis. N Engl J Med 378, 840–851. [DOI] [PubMed] [Google Scholar]
  9. Dalmau J, Tuzun E, Wu HY, Masjuan J, Rossi JE, Voloschin A, Baehring JM, Shimazaki H, Koide R, King D, et al. (2007). Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol 61, 25–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Emsley P, Lohkamp B, Scott WG, and Cowtan K (2010). Features and development of Coot. Acta Crystallogr D Biol Crystallogr 66, 486–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Gharpure A, Teng J, Zhuang Y, Noviello CM, Walsh RM Jr., Cabuco R, Howard RJ, Zaveri NT, Lindahl E, and Hibbs RE (2019). Agonist Selectivity and Ion Permeation in the alpha3beta4 Ganglionic Nicotinic Receptor. Neuron 104, 501–511 e506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Gielen M, and Corringer PJ (2018). The dual-gate model for pentameric ligand-gated ion channels activation and desensitization. J Physiol 596, 1873–1902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Hughes EG, Peng X, Gleichman AJ, Lai M, Zhou L, Tsou R, Parsons TD, Lynch DR, Dalmau J, and Balice-Gordon RJ (2010). Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci 30, 5866–5875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Jatczak-Sliwa M, Kisiel M, Czyzewska MM, Brodzki M, and Mozrzymas JW (2020). GABAA Receptor beta2E155 Residue Located at the Agonist-Binding Site Is Involved in the Receptor Gating. Front Cell Neurosci 14, 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Kawate T, and Gouaux E (2006). Fluorescence-detection size-exclusion chromatography for precrystallization screening of integral membrane proteins. Structure 14, 673–681. [DOI] [PubMed] [Google Scholar]
  16. Kim JJ, Gharpure A, Teng J, Zhuang Y, Howard RJ, Zhu S, Noviello CM, Walsh RM Jr., Lindahl E, and Hibbs RE (2020). Shared structural mechanisms of general anaesthetics and benzodiazepines. Nature 585, 303–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Kim JJ, and Hibbs RE (2021). Direct Structural Insights into GABAA Receptor Pharmacology. Trends Biochem Sci 46, 502–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Klesse G, Rao S, Sansom MSP, and Tucker SJ (2019). CHAP: A Versatile Tool for the Structural and Functional Annotation of Ion Channel Pores. J Mol Biol 431, 3353–3365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Kreye J, Wenke NK, Chayka M, Leubner J, Murugan R, Maier N, Jurek B, Ly LT, Brandl D, Rost BR, et al. (2016). Human cerebrospinal fluid monoclonal N-methyl-D-aspartate receptor autoantibodies are sufficient for encephalitis pathogenesis. Brain 139, 2641–2652. [DOI] [PubMed] [Google Scholar]
  20. Kreye J, Wright SK, van Casteren A, Stoffler L, Machule ML, Reincke SM, Nikolaus M, van Hoof S, Sanchez-Sendin E, Homeyer MA, et al. (2021). Encephalitis patient-derived monoclonal GABAA receptor antibodies cause epileptic seizures. J Exp Med 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Laverty D, Desai R, Uchanski T, Masiulis S, Stec WJ, Malinauskas T, Zivanov J, Pardon E, Steyaert J, Miller KW, et al. (2019). Cryo-EM structure of the human alpha1beta3gamma2 GABAA receptor in a lipid bilayer. Nature 565, 516–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Lefebvre SN, Taly A, Menny A, Medjebeur K, and Corringer PJ (2021). Mutational analysis to explore long-range allosteric couplings involved in a pentameric channel receptor pre-activation and activation. Elife 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Liebschner D, Afonine PV, Baker ML, Bunkoczi G, Chen VB, Croll TI, Hintze B, Hung LW, Jain S, McCoy AJ, et al. (2019). Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr D Struct Biol 75, 861–877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Lyons JA, Boggild A, Nissen P, and Frauenfeld J (2017). Saposin-Lipoprotein Scaffolds for Structure Determination of Membrane Transporters. Methods Enzymol 594, 85–99. [DOI] [PubMed] [Google Scholar]
  25. Masiulis S, Desai R, Uchanski T, Serna Martin I, Laverty D, Karia D, Malinauskas T, Zivanov J, Pardon E, Kotecha A, et al. (2019). GABAA receptor signalling mechanisms revealed by structural pharmacology. Nature 565, 454–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Mastronarde DN (2005). Automated electron microscope tomography using robust prediction of specimen movements. J Struct Biol 152, 36–51. [DOI] [PubMed] [Google Scholar]
  27. Morin A, Eisenbraun B, Key J, Sanschagrin PC, Timony MA, Ottaviano M, and Sliz P (2013). Collaboration gets the most out of software. Elife 2, e01456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Mortensen M, Iqbal F, Pandurangan AP, Hannan S, Huckvale R, Topf M, Baker JR, and Smart TG (2014). Photo-antagonism of the GABAA receptor. Nat Commun 5, 4454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Nemecz A, Prevost MS, Menny A, and Corringer PJ (2016). Emerging Molecular Mechanisms of Signal Transduction in Pentameric Ligand-Gated Ion Channels. Neuron 90, 452–470. [DOI] [PubMed] [Google Scholar]
  30. Newell JG, and Czajkowski C (2003). The GABAA receptor alpha 1 subunit Pro174-Asp191 segment is involved in GABA binding and channel gating. J Biol Chem 278, 13166–13172. [DOI] [PubMed] [Google Scholar]
  31. Newell JG, McDevitt RA, and Czajkowski C (2004). Mutation of glutamate 155 of the GABAA receptor beta2 subunit produces a spontaneously open channel: a trigger for channel activation. J Neurosci 24, 11226–11235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Nutt DJ, and Malizia AL (2001). New insights into the role of the GABA(A)-benzodiazepine receptor in psychiatric disorder. Br J Psychiatry 179, 390–396. [DOI] [PubMed] [Google Scholar]
  33. Obermayer-Straub P, Strassburg CP, and Manns MP (2000). Autoimmune hepatitis. J Hepatol 32, 181–197. [DOI] [PubMed] [Google Scholar]
  34. Petit-Pedrol M, Armangue T, Peng X, Bataller L, Cellucci T, Davis R, McCracken L, Martinez-Hernandez E, Mason WP, Kruer MC, et al. (2014). Encephalitis with refractory seizures, status epilepticus, and antibodies to the GABAA receptor: a case series, characterisation of the antigen, and analysis of the effects of antibodies. Lancet Neurol 13, 276–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, and Ferrin TE (2004). UCSF Chimera--a visualization system for exploratory research and analysis. J Comput Chem 25, 1605–1612. [DOI] [PubMed] [Google Scholar]
  36. Prüss H (2021). Autoantibodies in neurological disease. Nat Rev Immunol. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Reeves PJ, Callewaert N, Contreras R, and Khorana HG (2002). Structure and function in rhodopsin: high-level expression of rhodopsin with restricted and homogeneous N-glycosylation by a tetracycline-inducible N-acetylglucosaminyltransferase I-negative HEK293S stable mammalian cell line. Proc Natl Acad Sci U S A 99, 13419–13424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Reincke SM, Pruss H, and Kreye J (2020). Brain antibody sequence evaluation (BASE): an easy-to-use software for complete data analysis in single cell immunoglobulin cloning. BMC Bioinformatics 21, 446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Sigel E, and Ernst M (2018). The Benzodiazepine Binding Sites of GABAA Receptors. Trends Pharmacol Sci 39, 659–671. [DOI] [PubMed] [Google Scholar]
  40. Smart OS, Neduvelil JG, Wang X, Wallace BA, and Sansom MS (1996). HOLE: a program for the analysis of the pore dimensions of ion channel structural models. J Mol Graph 14, 354–360, 376. [DOI] [PubMed] [Google Scholar]
  41. Tonon MC, Vaudry H, Chuquet J, Guillebaud F, Fan J, Masmoudi-Kouki O, Vaudry D, Lanfray D, Morin F, Prevot V, et al. (2020). Endozepines and their receptors: Structure, functions and pathophysiological significance. Pharmacol Ther 208, 107386. [DOI] [PubMed] [Google Scholar]
  42. Vaughn JL, Goodwin RH, Tompkins GJ, and McCawley P (1977). The establishment of two cell lines from the insect Spodoptera frugiperda (Lepidoptera; Noctuidae). In Vitro 13, 213–217. [DOI] [PubMed] [Google Scholar]
  43. Wagner T, Merino F, Stabrin M, Moriya T, Antoni C, Apelbaum A, Hagel P, Sitsel O, Raisch T, Prumbaum D, et al. (2019). SPHIRE-crYOLO is a fast and accurate fully automated particle picker for cryo-EM. Commun Biol 2, 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Waterhouse A, Bertoni M, Bienert S, Studer G, Tauriello G, Gumienny R, Heer FT, de Beer TAP, Rempfer C, Bordoli L, et al. (2018). SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res 46, W296–W303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Williams CJ, Headd JJ, Moriarty NW, Prisant MG, Videau LL, Deis LN, Verma V, Keedy DA, Hintze BJ, Chen VB, et al. (2018). MolProbity: More and better reference data for improved all-atom structure validation. Protein Sci 27, 293–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Zhang K (2016). Gctf: Real-time CTF determination and correction. J Struct Biol 193, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Zheng SQ, Palovcak E, Armache JP, Verba KA, Cheng Y, and Agard DA (2017). MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat Methods 14, 331–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Zhu S, Noviello CM, Teng J, Walsh RM Jr., Kim JJ, and Hibbs RE (2018). Structure of a human synaptic GABAA receptor. Nature 559, 67–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Zivanov J, Nakane T, Forsberg BO, Kimanius D, Hagen WJ, Lindahl E, and Scheres SH (2018). New tools for automated high-resolution cryo-EM structure determination in RELION-3. Elife 7. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

Supplemental Figure 1: Patient-derived antibodies bind to the α1β2γ2 GABAA receptor and representative purification, related to Figure 1. A, Purified GABAA receptor was reconstituted in nanodiscs in the absence or presence of each monoclonal antibody (mAb) indicated and analyzed by fluorescence-detection size exclusion chromatography (FSEC). A shift to the left indicates an increase in size associated with binding. B-E are screens of selected Fab fragments produced from the mAbs. Increasing amounts of Fab were added to GABAA receptor in nanodiscs to determine at which point the elution volume shift is saturated. F, Immunofluorescence staining of selected GABAA Fabs (green) on PFA-fixed murine brain tissue sections shown in the CA3 region of the hippocampus with DRAQ5 stained nuclei (red). Representative scale bar indicates 50 μm. G, FSEC traces of receptor alone (black trace) and receptor plus Fab115 (blue trace). H, FSEC trace of the final material applied to EM grids; inset shows Coomassie-stained SDS-PAGE gel of the complex.

2

Supplemental Figure 2: Cryo-EM processing workflow for Fab175 + GABAA, related to Figures 1, 3. Each panel represents a different stage of the data collection and processing. A, Representative micrograph. Scale bar indicates 100 nm. B, 2D classification with selected classes boxed in red. C, First 3D classification with selected class indicated. D, First refinement. E, Second 3D classification using the refined angles from D to perform fine local angular searches. F, Second refinement. G, 3D classification on signal subtracted particles to focus alignment on the γ-TMD. H, Final refinement, sharpening, local resolution estimation and FSC Curve for the Fab175+GABAA dataset. I, as in H but for the Fab115+GABAA dataset.

3

Supplemental Figure 3: Quality of Cryo-EM maps at regions of interaction between Fab and receptor, related to Figures 2, 3. A, Overview of Fab115:GABAA complex map from view parallel to membrane. Panel D inset is indicated. B, As in panel A, but turned 180° to show other Fab115:GABAA interface. Inset indicated is shown in panel E. Mesh in panels D and E is the result of high-pass filtering the density maps to 6 Å in order to emphasize higher resolution features. C, Overview of Fab115:GABAA complex map from view of outside the cell looking in. Panel F inset is indicated, highlighting the interaction between the Fab CDR3 and neurotransmitter binding domain. G, Overview of Fab175:GABAA complex map from view parallel to membrane. Panel I inset is indicated. H, Overview of Fab175:GABAA complex map from view of outside the cell looking in. Panel J inset is indicated, highlighting CDR3 of the antibody heavy chain, which inserts into the ECD α1:γ2 interface.

4

Supplemental Figure 4: Overview of receptor:Fab models, related to Figures 14. A, Fab115: GABAA model. ECD, extracellular domain. TMD, transmembrane domain. B, Fab115: HC, heavy chain; LC, light chain. CDR, complementarity determining region. The CDRs are colored in red-orange for contrast. C, D Same as for A, B but CDRs are colored green. E, F Subunit interface contacts with Fab. E, One β2:α1 subunit interface is shown; red box indicates neurotransmitter binding site and spheres indicate amino acids contacted by Fab115. Inset table shows surface area buried by Fab heavy and light chains on respective GABAA subunits. F, Same as for E but for Fab175.

5

Supplemental Figure 5: Alignment of GABAA receptor subunits contacted by autoantibodies, related to Figures 23. Amino acid sequence alignments of GABAA subunits contacted by Fab115 and Fab175 are shown. On the right-hand side of the figure are cartoons indicating the location of the antibodies bound (yellow is Fab115, red is Fab175). Subunits shown in the alignment for each box are colored.

6

Supplemental Figure 6: Somatic hypermutations of Fab115 and Fab175 compared to receptor contacts, related to Figures 23. A, Alignments between sequences of Fab115 and its presumed germline ancestor. Only the variable domain is shown. CDR1, CDR2, and CDR3 are indicated in bold for each antibody segment. Somatic hypermutations are indicated by noting the parental sequence in the germline. Residues of the V(D)J junctions are underlined. Note, these residues cannot reliably be differentiated from germline ancestors. Hence assignment of somatic hypermutations is limited to V and J genes only, and thus not for most part of the highly variable CDR3. Residues of the heavy and light chains that contact the GABAA receptor are indicated and color coded based on subunit. The NG-KR mutations discussed in the text are indicated by asterisks (*). B, Same as in A but for Fab175.

7

Supplemental Table 1: Cryo-EM Data Collection, Processing and Model Statistics, refers to Figures 1, S1, S2.

8

Supplemental Video 1: Overview and zooms of Fab115:GABAA map density, modeling, and quality, refers to Figures 12, S1S3

Download video file (119.5MB, mp4)
9

Supplemental Video 2: Overview and zooms of Fab175:GABAA map density, modeling, and quality, refers to Figures 1, 3, S1S3

Download video file (54.5MB, mp4)

Data Availability Statement

  • Cryo-EM maps and atomic model coordinates have been deposited in the EMDB and RCSB, respectively, under the accession codes 7T0W/EMD-25583 and 7T0Z/EMD-25585. They will be publicly available as of the date of publication.

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

RESOURCES