Protein instability, haploinsufficiency, and cortical hyper-excitability underlie STXBP1 encephalopathy

Jovana Kovacevic, Gregoire Maroteaux, Desiree Schut, Maarten Loos, Mohit Dubey, Julika Pitsch, Esther Remmelink, Bastijn Koopmans, James Crowley, L Niels Cornelisse, Patrick F Sullivan, Susanne Schoch, Ruud F Toonen, Oliver Stiedl, Matthijs Verhage, Jovana Kovacevic, Gregoire Maroteaux, Desiree Schut, Maarten Loos, Mohit Dubey, Julika Pitsch, Esther Remmelink, Bastijn Koopmans, James Crowley, L Niels Cornelisse, Patrick F Sullivan, Susanne Schoch, Ruud F Toonen, Oliver Stiedl, Matthijs Verhage

Abstract

De novo heterozygous mutations in STXBP1/Munc18-1 cause early infantile epileptic encephalopathies (EIEE4, OMIM #612164) characterized by infantile epilepsy, developmental delay, intellectual disability, and can include autistic features. We characterized the cellular deficits for an allelic series of seven STXBP1 mutations and developed four mouse models that recapitulate the abnormal EEG activity and cognitive aspects of human STXBP1-encephalopathy. Disease-causing STXBP1 variants supported synaptic transmission to a variable extent on a null background, but had no effect when overexpressed on a heterozygous background. All disease variants had severely decreased protein levels. Together, these cellular studies suggest that impaired protein stability and STXBP1 haploinsufficiency explain STXBP1-encephalopathy and that, therefore, Stxbp1+/- mice provide a valid mouse model. Simultaneous video and EEG recordings revealed that Stxbp1+/- mice with different genomic backgrounds recapitulate the seizure/spasm phenotype observed in humans, characterized by myoclonic jerks and spike-wave discharges that were suppressed by the antiepileptic drug levetiracetam. Mice heterozygous for Stxbp1 in GABAergic neurons only, showed impaired viability, 50% died within 2-3 weeks, and the rest showed stronger epileptic activity. c-Fos staining implicated neocortical areas, but not other brain regions, as the seizure foci. Stxbp1+/- mice showed impaired cognitive performance, hyperactivity and anxiety-like behaviour, without altered social behaviour. Taken together, these data demonstrate the construct, face and predictive validity of Stxbp1+/- mice and point to protein instability, haploinsufficiency and imbalanced excitation in neocortex, as the underlying mechanism of STXBP1-encephalopathy. The mouse models reported here are valid models for development of therapeutic interventions targeting STXBP1-encephalopathy.

Figures

Figure 1
Figure 1
Morphological and electrophysiological characteristics of dissociated hippocampal neurons expressing the human disease variants in Stxbp1 null mouse neurons. (A) Schematic overview of some of the previously discovered STXBP1 truncations, deletions and missense mutations in human patients showing the seven mutations tested in colour-coded boxes. (B) Dissociated cortical neurons were stained for Munc18-1, dendritic marker MAP2 and synaptic marker synaptobrevin (VAMP). Examples represent Stxbp1 null neurons expressing wild-type Munc18-1 (WT), C180Y or M433R. (C–G) Morphological and synaptic characteristics of Munc18-1 wild-type neurons, Stxbp1+/− neurons and null mutant neurons expressing one of the human disease variants: (C) mean synapse density calculated as the ratio of synapse number and total dendritic length. (D) Mean soma Munc18-1 level. (E) Mean synaptic Munc18-1 level. (F and G) Mean somatic and synaptic syntaxin-1 level. (H) Example traces of evoked release from wild-type neurons, Stxbp1 null neurons expressing C180Y and M433R upon a single action potential stimulation. (I) Mean EPSC amplitude expressed as the ratio of the wild-type value. (J) Paired-pulse ratio (calculated as the ratio of second EPSC and first EPSC) depending on the pulse interval. (K) Frequency of spontaneous release normalized to the wild-type values. (L). Amplitude of spontaneous release normalized to the wild-type values. (M) Example traces of spontaneous release in Stxbp1 null neurons expressing Munc18-1 wild-type, C180Y or M433R human variants. (N) The size of the readily releasable pool derived from back extrapolation of cumulative total charge released during 40 Hz train, 100 APs. The number of analysed cells is indicated in the bars. *P < 0.05, **P < 0.01, ***P < 0.001 versus infected wild-type control. Explanation of statistical analysis is provided in the Supplementary material. mEPSC = mini excitatory postsynaptic current; RRP = readily releasable pool.
Figure 2
Figure 2
Morphological and electrophysiological characteristics of dissociated hippocampal neurons expressing the human disease variants in Stxbp1+/− mouse neurons. (A) Dissociated cortical neurons were stained for Munc18-1 and synaptic marker synaptobrevin (VAMP). Examples represent Stxbp1+/− neurons expressing wild-type Munc18-1 (WT), C180Y or M433R human variants. (B–D) Morphological and synaptic characteristics of Stxbp1+/− neurons expressing wild-type Munc18-1 or one of the human disease variants: (B) mean synapse density: calculated as the ratio of synapse number and total dendritic length. (C and D) Mean somatic and synaptic Munc18-1 level. (E) Example traces of evoked release upon a single action potential stimulation from Stxbp1+/− neurons expressing wild-type Munc18-1, C180Y, M433R, V84D, G544D or R388X human variants. (F) Mean EPSC amplitude expressed as the ratio of the infected wild-type values. (G) Paired-pulse ratio (second EPSC/first EPSC) depending on the pulse interval. (H) Readily releasable pool estimate derived from back-extrapolation of cumulative total charge released during 40 Hz train, 100 APs (I) Frequency of spontaneous release expressed as the ratio of infected wild-type values. (J) Amplitude of spontaneous release expressed as the ratio of infected wild-type values. (K) Example traces of spontaneous release in Stxbp1+/− neurons expressing wild-type Munc18-1, C180Y, M433R, G544D, R388X or V84D human variants. The number of analysed cells is indicated in the bars. ***P < 0.001 versus infected wild-type control. mEPSC = mini excitatory postsynaptic current; RRP = readily releasable pool.
Figure 3
Figure 3
Cellular stability of wild-type and human disease variants of Munc18-1 in HEK293 cells and neurons. (A) Immunochemistry of HEK293 cells infected with wild-type Munc18-1 (WT), C180Y, M443R, C522R and T574P constructs stained for Munc18-1, EGFP and Golgi marker (GM130). (B) Normalized Munc18-1 levels in HEK293 cells after viral infection with wild-type, C180Y, M443R, C522R and T574P constructs. The inset shows representative western blot of HEK293 cells after viral infection; n = 5, 5, 5, 2 and 2, respectively. (C) Western blot analysis of Munc18-1 protein levels 0, 6, 12, 24 and 30 h after block of protein synthesis with cycloheximide for HEK293 cells infected with wild-type, C180Y, M433R, C522R or T574P constructs. The infection with wild-type construct was used as a control for all performed western blot analysis. (D) Quantitative analysis of the Munc18-1 protein expression from western blots in HEK cells represented in C. (E) Western blot analysis of Munc18-1 protein levels 0, 12, 24 and 36 h after block of protein synthesis with cycloheximide for wild-type, C180Y, M433R, C522R or T574P constructs in Stxbp1 null neurons. The infection with wild-type construct was used as a control for all performed western blot analysis. (F) Quantitative analysis of the Munc18-1 protein expression from western blots in neurons represented in E. (G–I) Normalized Munc18-1 protein levels from three constructs expressed in HEK cells treated with MG132, Leupeptin or Pepstatin; n = 3, 2 and 2, respectively.
Figure 4
Figure 4
Generation, genomic analysis, routine behavioural observation and spontaneous activity of Stxbp1+/− mice. (A–D) Generations of four lines of Stxbp1+/− mice: conditional, congenic BL6, reverse 129Sv and Gad2- Stxbp1 line. Chromosomes bearing Stxbp1 mutation originating from the C57BL/6J genetic background are shown in grey, and those from the 129/SvJ genetic background are shown in orange. Stxbp1 mutation is represented with grey triangle; LoxP sites are represented with black rectangles and Cre-deleter lines are represented with scissors (red scissors: Cre expressed in all neuron, blue scissors: Gad2tm2-Cre mice with Cre-recombinase expressed in only GABAergic neurons). The flanking gene region is represented as orange region in the grey chromosome in the congenic BL6 line and vice versa in the reverse 129Sv line (adapted from Wolfer et al., 2002). (E and F) High-density genomic analysis showed the size and position of flanking gene region in Stxbp1+/− samples from congenic BL6 and reverse 129Sv lines. ‘Me-PaMuFind-It’ web tool (http://me-pamufind-it.org/) revealed three genes with passenger mutations from 129Sv genetic background within flanking genes region in congenic BL6 Stxbp1+/− samples. (G) Stxbp1+/− mice showed lower body weight compared to their controls. (H) Grip strength was normal in Stxbp1+/− mice. (I) Motor coordination and motor learning in Stxbp1+/− mice was normal, as assessed on the rotarod. (J and K) Circadian rhythm assessed by changes in the activity in anticipation of (filled bars), and response to (open bars), day/night transitions and proportion of time spent outside the shelter was normal in Stxbp1+/− mice. (L) Proportion of activity duration during the first 3 h of the dark phase days in the home-cage environment (PhenoTyper) showed increased activity of Stxbp1+/− mice from all three lines during the first dark phase. (M) Proportion of activity duration in the PhenoTyper during the light phases was overall lower for Stxbp1+/− mice (conditional mice, congenic BL6 and reverse 129Sv). The number of animals assigned is shown in the graphs. *P < 0.05; ***P < 0.001.
Figure 5
Figure 5
Video and EEG recordings revealed epileptic-like events in Stxbp1+/− mice. (A) Video monitoring revealed sudden jerks referred as twitches in Stxbp1+/− mice. (B) Video monitoring revealed sudden jumps in Stxbp1+/− mice, sometimes accompanied by Straub tail responses previously reported as a common phenomenon observed after seizure onset (Wagnon et al., 2015). (C) Distribution of motor effects of epileptic-like neural activity (twitches and jumps) during 3-h video-monitoring in Stxbp1+/− mice from three lines: floxed, congenic BL6 and reverse 129Sv line. These motor events were never found in control mice (Stxbp1+/+). (D) Monitoring for 24 h showed that most of the twitches and jumps in congenic BL6 Stxbp1+/− mouse occurred during the light phase (twitch: 22/34 and jump: 2/3) of the day/night cycle. (E) Average number of behavioural epileptic-like events per hour per line of Stxbp1+/− mice. (F) Positions of the recording electrodes and ground electrode relative to Bregma. (G) Representative example of ECoG traces in a congenic BL6 Stxbp1+/− mouse during the slow-wave sleep, awake state and spike-wave discharges. The red trace is an expanded ECoG trace of spike-wave discharge. (H) Power spectrum of slow wave sleep (SWS), spike wave discharge (SWD) and wake state. (I) Occurrence of behavioural epileptic events (twitches and jumps) and spike-wave discharges detected in cortical and hippocampal EEG traces during 3-h recording in Stxbp1cre/+ and congenic BL6 Stxbp1+/− mice. (J) Predicted probability of coincidence of 3/13 twitches and 1/7 jumps with spike-wave discharge detected in a representative congenic BL6 Stxbp1+/− mouse. Probability lower than 5% (grey line) was considered as concurrence. (K) Probability of concurrence of behavioural epileptic events and spike-wave discharges within 10 s presented as a negative logarithm for Stxbp1cre/+ and congenic BL6 Stxbp1+/− mice. (L) Total number of detected spike-wave discharges and epileptic events in the 12-h light phase. The number of mice: three Stxbp1cre/+ mice, four congenic BL6 Stxbp1+/− mice for cortical recording and two congenic BL6 Stxbp1+/− mice for hippocampal recording. (M) Average frequency of detected spike-wave discharges during 6 h of recording after administration of saline, first levetiracetam dose (LEV I: 50 mg/kg, i.p.) and fifth levetiracetam dose (LEV V: 5 days, 50 mg/kg per day, i.p.). Different greyscale circles represent individual mice. *P < 0.05, **P < 0.01 compared to saline administration.
Figure 6
Figure 6
c-Fos expression in Stxbp1+/− mice. (AR) Representatives of c-Fos expression in prefrontal cortex (PFC), primary motor cortex (mCx) and somatosensory cortex (ssCx) for Stxbp1+/+ mice (control), Stxbp1cre/+ and congenic BL6 Stxbp1+/− mice. (S) Number of c-Fos positive cells per brain region for Stxbp1+/+, Stxbp1cre/+, and congenic BL6 Stxbp1+/− mice. Scale bars and the number of samples are provided in the figure.
Figure 7
Figure 7
Learning and memory in Stxbp1+/− mice in classical spatial paradigms and recently developed automated tasks in the PhenoTyper. (A and B) Latency and distance travelled to find the escape hole in the Barnes maze during the learning phase for congenic BL6 Stxbp1+/− mice. Congenic BL6 Stxbp1+/− (HZ BL6) showed longer latency to find new escape hole during the learning phase (P = 0.026) and during R1 (P < 0.001) and travelled longer distance to the new escape hole (R1-R3: P = 0.024) compared to their controls (WT BL6). (C) HZ BL6 mice showed narrower distribution of holes visit around the target hole during the first probe trial (P1) compared to wild-type BL6. (D) Probability of hole visits in the old target octant during the P1 and P2 tended to be higher for HZ BL6 mice compared to their controls (P = 0.086 and P = 0.060, respectively) and there were no differences in the probability of hole visits in the new target octant during the P2. (E and F) Latency and distance travelled to find the escape hole in the Barnes maze during the learning phase were similar for Stxbp1cre/+ mice (HZ cond) and their controls (wild-type cond). (G and H) Escape latency and distance travelled to the platform during the training in the Morris water maze was similar for reverse 129Sv Stxbp1+/− mice and control mice. (I) Time spent per quadrant during the probe trial was similar for reverse 129Sv Stxbp1+/− mice and control mice. (J) Schematic overview of the CognitionWall DL/RL task. (K and L) Kaplan-Meier survival curves shows the fraction of congenic BL6 and conditional Stxbp1 mice that reached the 80% criterion as a function of hole entries during the DL and RL phases. (M) Average number of entries made per group to reach 80% criterion during the DL and RL phases. HZ BL6 mice reached the 80% criterion during RL with lower number of entries compared to control (P = 0.004). (N) Schematic overview of the Shelter task protocol in the PhenoTyper to assess avoidance learning. (O and P) The preference index during the dark phases of the avoidance learning task was similar for HZ BL6 and HZ cond mice and their controls. Insets represent the learning effect on the preference index (D5/D6/D7-D4). The insets in graph O shows that congenic BL6 Stxbp1+/− mice showed a stronger learning effect compared to their controls (P = 0.012). (Q and R) The aversion index during the dark phases of avoidance learning task showed trend toward lower values for HZ BL6 mice compared to their controls (P = 0.101). The aversion index was similar between HZ cond mice and their controls. *P < 0.05; **P < 0.01; ***P < 0.001 compared to respective control. Statistical analysis is explained in the Supplementary material.
Figure 8
Figure 8
Anxiety-related phenotype and social behaviour in Stxbp1+/− mice. (A) Schematic overview of the PhenoTyper home-cage. (B) On shelter duration during the dark phase was shorter for Stxbp1cre/+ and congenic BL6 Stxbp1+/− (HZ cond and HZ BL6) mice compared to their controls. (C–F) Elevated plus maze (EPM) data. (C) Schematic overview of the EPM. (D–F) Latency to enter open arms was longer, time spent on the open arms and number of visits to the open arms were lower for Stxbp1cre/+ and congenic BL6 Stxbp1+/− mice compared to their controls. (G–J) Dark-light box (DLB) data. (G) Schematic overview of the DLB. (H) Latency to visit bright compartment (BC) was longer for congenic BL6 Stxbp1+/− mice compared to their controls. (I–J) Time spent in the bright compartment and number of visits to the bright compartment was lower for Stxbp1cre/+ and congenic BL6 Stxbp1+/− mice compared to their controls. (K–O) Open field data. (K) Schematic overview of the open field. (L) Latency to visit centre zone was longer for Stxbp1cre/+ and congenic BL- Stxbp1+/− mice compared to their controls. (M and N) Percentage of distance moved in the centre zone and number of visits to centre zone was similar for Stxbp1cre/+ and congenic BL6 Stxbp1+/− mice and respective control mice. (O) The total distance moved was longer for reverse 129Sv Stxbp1+/− mice compared to their control, and it was similar for Stxbp1cre/+ and congenic BL6 Stxbp1+/− mice and respective control mice. (P and Q) Novelty induced hypophagia test (NIHP) data. (P) Schematic overview of the NIHP test. (Q) Latency to consume highly palatable food (cracker) was similar for Stxbp1cre/+ and congenic BL6 Stxbp1+/− mice and the respective controls. (R) Schematic overview of three-chamber test protocol. (S) During test for sociability (phase III), Stxbp1cre/+, congenic BL6 Stxbp1+/− mice and their respective controls spent more time in the mouse zone than in the object containing zone. (T) During test for social novelty (phase IV), Stxbp1cre/+, congenic BL6 Stxbp1+/− mice and their respective controls spent more time in the zone with novel mouse than in the zone with familiar mouse. There were no differences between genotypes. *P < 0.05, **P < 0.01, ***P < 0.001 for comparison between zones.

References

    1. Barcia G, Chemaly N, Gobin S, Milh M, Van Bogaert P, Barnerias C, et al.Early epileptic encephalopathies associated with STXBP1 mutations: could we better delineate the phenotype? Eur J Med Genet 2014; 57: 15–20.
    1. Beyenburg S, Mitchell AJ, Schmidt D, Elger CE, Reuber M. Anxiety in patients with epilepsy: systematic review and suggestions for clinical management [Review]. Epilepsy Behav 2005; 7: 161–71.
    1. Brenner S. The genetics of Caenorhabditis elegans. Genetics 1974; 77: 71–94.
    1. Brose N, O'Connor V, Skehel P. Synaptopathy: dysfunction of synaptic function? Biochem Soc Trans 2010; 38: 443–4.
    1. Campbell IM, Yatsenko SA, Hixson P, Reimschisel T, Thomas M, Wilson W, et al.Novel 9q34.11 gene deletions encompassing combinations of four Mendelian disease genes: STXBP1, SPTAN1, ENG, and TOR1A. Genet Med 2012; 14: 868–76.
    1. Cerqueira JJ, Mailliet F, Almeida OF, Jay TM, Sousa N. The prefrontal cortex as a key target of the maladaptive response to stress. J Neurosci 2007; 27: 2781–7.
    1. Chai YJ, Sierecki E, Tomatis VM, Gormal RS, Giles N, Morrow IC, et al.Munc18-1 is a molecular chaperone for α-synuclein, controlling its self-replicating aggregation. J Cell Biol 2016; 214: 705–18.
    1. Corradini I, Donzelli A, Antonucci F, Welzl H, Loos M, Martucci R, et al.Epileptiform activity and cognitive deficits in SNAP-25(+/-) mice are normalized by antiepileptic drugs. Cereb Cortex 2014; 24: 364–76.
    1. Crawley JN. Behavioral phenotyping strategies for mutant mice. Neuron 2008; 57: 809–18.
    1. Crunelli V, Leresche N. Childhood absence epilepsy: genes, channels, neurons and networks [Review]. Nat Rev Neurosci 2002; 3: 371–82.
    1. de Rubeis S, He X, Goldberg AP, Poultney CS, Samocha K, Cicek AE, et al.Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 2014; 515: 209–15.
    1. de Vries KJ, Geijtenbeek A, Brian EC, de Graan PN, Ghijsen WE, Verhage M. Dynamics of munc18-1 phosphorylation/dephosphorylation in rat brain nerve terminals. Eur J Neurosci 2000; 12: 385–90.
    1. Deprez L, Weckhuysen S, Holmgren P, Suls A, Van Dyck T, Goossens D, et al.Clinical spectrum of early-onset epileptic encephalopathies associated with STXBP1 mutations. Neurology 2010; 75: 1159–65.
    1. Dias-Ferreira E, Sousa JC, Melo I, Morgado P, Mesquita AR, Cerqueira JJ, et al.Chronic stress causes frontostriatal reorganization and affects decision-making. Science 2009; 325: 621–5.
    1. Dilena R, Striano P, Traverso M, Viri M, Cristofori G, Tadini L, et al.Dramatic effect of levetiracetam in early-onset epileptic encephalopathy due to STXBP1 mutation. Brain Dev 2016; 38: 128–31.
    1. Doheny HC, Ratnaraj N, Whittington MA, Jefferys JG, Patsalos PN. Blood and cerebrospinal fluid pharmacokinetics of the novel anticonvulsant levetiracetam (ucb L059) in the rat. Epilepsy Res 1999; 34: 161–8.
    1. Fassio A, Patry L, Congia S, Onofri F, Piton A, Gauthier J, et al.SYN1 loss-of-function mutations in autism and partial epilepsy cause impaired synaptic function. Hum Mol Genet 2011; 20: 2297–307.
    1. Garcia CC, Blair HJ, Seager M, Coulthard A, Tennant S, Buddles M, et al.Identification of a mutation in synapsin I, a synaptic vesicle protein, in a family with epilepsy. J Med Genet 2004; 41: 183–6.
    1. Gerber SH, Rah JC, Min SW, Liu X, de Wit H, Dulubova I, et al.Conformational switch of syntaxin-1 controls synaptic vesicle fusion. Science 2008; 321: 1507–10.
    1. Gerlai R. Gene targeting: technical confounds and potential solutions in behavioral brain research [Review]. Behav Brain Res 2001; 125: 13–21.
    1. Geschwind DH, Levitt P. Autism spectrum disorders: developmental disconnection syndromes. Curr Opin Neurobiol 2007; 17: 103–11.
    1. Graybeal C, Feyder M, Schulman E, Saksida LM, Bussey TJ, Brigman JL, et al.Paradoxical reversal learning enhancement by stress or prefrontal cortical damage: rescue with BDNF. Nat Neurosci 2011; 14: 1507–9.
    1. Graybeal C, Bachu M, Mozhui K, Saksida LM, Bussey TJ, Sagalyn E, et al.Strains and stressors: an analysis of touchscreen learning in genetically diverse mouse strains. PLoS One 2014; 9: e87745.
    1. Grone BP, Marchese M, Hamling KR, Kumar MG, Krasniak CS, Sicca F, et al.Epilepsy, behavioral abnormalities, and physiological comorbidities in syntaxin-binding protein 1 (STXBP1) mutant Zebrafish. PLoS One 2016; 11: e0151148.
    1. Guacci A, Chetta M, Rizzo F, Marchese G, De Filippo MR, Giurato G, et al.Phenytoin neurotoxicity in a child carrying new STXBP1 and CYP2C9 gene mutations. Seizure 2016; 34: 26–8.
    1. Hager T, Maroteaux G, du Pont P, Julsing J, van Vliet R, Stiedl O. Munc18-1 haploinsufficiency results in enhanced anxiety-like behavior as determined by heart rate responses in mice. Behav Brain Res 2014; 260: 44–52.
    1. Hamdan FF, Piton A, Gauthier J, Lortie A, Dubeau F, Dobrzeniecka S, et novo STXBP1 mutations in mental retardation and nonsyndromic epilepsy. Ann Neurol 2009; 65: 748–53.
    1. Hamdan FF, Gauthier J, Dobrzeniecka S, Lortie A, Mottron L, Vanasse M, et al.Intellectual disability without epilepsy associated with STXBP1 disruption. Eur J Hum Genet 2011; 19: 607–9.
    1. Han S, Tai C, Westenbroek RE, Yu FH, Cheah CS, Potter GB, et al.Autistic-like behaviour in Scn1a+/- mice and rescue by enhanced GABA-mediated neurotransmission. Nature 2012; 489: 385–90.
    1. Harrison FE, Hosseini AH, McDonald MP. Endogenous anxiety and stress responses in water maze and Barnes maze spatial memory tasks. Behav Brain Res 2009; 198: 247–51.
    1. Harrison SD, Broadie K, van de Goor J, Rubin GM. Mutations in the Drosophila Rop gene suggest a function in general secretion and synaptic transmission. Neuron 1994; 13: 555–66.
    1. Hata Y, Südhof TC. A novel ubiquitous form of Munc-18 interacts with multiple syntaxins. Use of the yeast two-hybrid system to study interactions between proteins involved in membrane traffic. J Biol Chem 1995; 270: 13022–8.
    1. Heeroma JH, Roelandse M, Wierda K, van Aerde KI, Toonen RF, Hensbroek RA, et al.Trophic support delays but does not prevent cell-intrinsic degeneration of neurons deficient for munc18-1. Eur J Neurosci 2004; 20: 623–34.
    1. Herrera DG, Robertson HA. Activation of c-fos in the brain [Review]. Prog Neurobiol 1996; 50: 83–107.
    1. Holmes A, Wrenn CC, Harris AP, Thayer KE, Crawley JN. Behavioral profiles of inbred strains on novel olfactory, spatial and emotional tests for reference memory in mice [Review]. Genes Brain Behav 2002; 1: 55–69.
    1. Homanics GE, Quinlan JJ, Firestone LL. Pharmacologic and behavioral responses of inbred C57BL/6J and strain 129/SvJ mouse lines. Pharmacol Biochem Behav 1999; 63: 21–6.
    1. Kataoka M, Yamamori S, Suzuki E, Watanabe S, Sato T, Miyaoka H, et al.A single amino acid mutation in SNAP-25 induces anxiety-related behavior in mouse. PLoS One 2011; 6: e25158.
    1. Kitamura K, Itou Y, Yanazawa M, Ohsawa M, Suzuki-Migishima R, Umeki Y, et al.Three human ARX mutations cause the lissencephaly-like and mental retardation with epilepsy-like pleiotropic phenotypes in mice. Hum Mol Genet 2009; 18: 3708–24.
    1. Keogh MJ, Daud D, Pyle A, Duff J, Griffin H, He L, et al.A novel de novo STXBP1 mutation is associated with mitochondrial complex I deficiency and late-onset juvenile-onset parkinsonism. Neurogenetics 2015; 16: 65–7.
    1. Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, et al.Analysis of protein-coding genetic variation in 60,706 humans. Nature 2016; 536: 285–91.
    1. Li L, Chin LS, Shupliakov O, Brodin L, Sihra TS, Hvalby O, et al.Impairment of synaptic vesicle clustering and of synaptic transmission, and increased seizure propensity, in synapsin I-deficient mice. Proc Natl Acad Sci USA 1995; 92: 9235–9.
    1. Loos M, Koopmans B, Aarts E, Maroteaux G, van der Sluis S, Neuro-BSIK Mouse Phenomics Consortium, et al.Sheltering behavior and locomotor activity in 11 genetically diverse common inbred mouse strains using home-cage monitoring. PLoS One 2014; 9: e108563.
    1. Löscher W, Hönack D. Profile of ucb L059, a novel anticonvulsant drug, in models of partial and generalized epilepsy in mice and rats. Eur J Pharmacol 1993; 232: 147–58.
    1. Lynch BA, Lambeng N, Nocka K, Kensel-Hammes P, Bajjalieh SM, Matagne A, et al.The synaptic vesicle protein SV2A is the binding site for the antiepileptic drug levetiracetam. Proc Natl Acad Sci USA 2004; 101: 9861–6.
    1. Maroteaux G, Loos M, van der Sluis S, Koopmans B, Aarts E, van Gassen K, et al.High-throughput phenotyping of avoidance learning in mice discriminates different genotypes and identifies a novel gene. Genes Brain Behav 2012; 11: 772–84.
    1. Martin S, Papadopulos A, Tomatis VM, Sierecki E, Malintan NT, Gormal RS, et al.Increased polyubiquitination and proteasomal degradation of a Munc18-1 disease-linked mutant causes temperature-sensitive defect in exocytosis. Cell Rep 2014; 9: 206–18.
    1. Meijer M, Burkhardt P, de Wit H, Toonen RF, Fasshauer D, Verhage M. Munc18-1 mutations that strongly impair SNARE-complex binding support normal synaptic transmission. EMBO J 2012; 31: 2156–68.
    1. Mignot C, Moutard ML, Trouillard O, Gourfinkel-An I, Jacquette A, Arveiler B, et al.STXBP1-related encephalopathy presenting as infantile spasms and generalized tremor in three patients. Epilepsia 2011; 52: 1820–7.
    1. Milh M, Villeneuve N, Chouchane M, Kaminska A, Laroche C, Barthez MA, et al.Epileptic and nonepileptic features in patients with early onset epileptic encephalopathy and STXBP1 mutations. Epilepsia 2011; 52: 1828–34.
    1. Misura KM, Scheller RH, Weis WI. Three-dimensional structure of the neuronal-Sec1-syntaxin 1a complex. Nature 2000; 404: 355–62.
    1. Nguyen HT, Bryois J, Kim A, Dobbyn A, Huckins LM, Munoz-Manchado A, et al.Integrated Bayesian analysis of rare exonic variants to identify risk genes for schizophrenia and neurodevelopmental disorders. Genome Med 2017; 9: 114 doi: 10.1101/135293.
    1. Onat FY, van Luijtelaar G, Nehlig A, Snead OC 3rd. The involvement of limbic structures in typical and atypical absence epilepsy. Epilepsy Res 2013; 103: 111–23.
    1. Otsuka M, Oguni H, Liang JS, Ikeda H, Imai K, Hirasawa K, et al.STXBP1 mutations cause not only Ohtahara syndrome but also West syndrome—result of Japanese cohort study. Epilepsia 2010; 51: 2449–52.
    1. Paxinos G, Franklin K. The mouse brain in stereotaxic coordinates. San Diego: Academic Press; 2012.
    1. Peñagarikano O, Abrahams BS, Herman EI, Winden KD, Gdalyahu A, Dong H, et al.Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities, and core autism-related deficits. Cell 2011; 147: 235–46.
    1. Pitsch J, Becker AJ, Schoch S, Müller JA, de Curtis M, Gnatkovsky V. Circadian clustering of spontaneous epileptic seizures emerges after pilocarpine-induced status epilepticus. Epilepsia 2017; 58: 1159–71.
    1. Remmelink E, Smit AB, Verhage M, Loos M. Measuring discrimination- and reversal learning in mouse models within 4 days and without prior food deprivation. Learn Mem 2016; 23: 660–7.
    1. Rohena L, Neidich J, Truitt Cho M, Gonzalez KD, Tang S, Devinsky O, Chung WK. Mutation in SNAP25 as a novel genetic cause of epilepsy and intellectual disability. Rare Dis 2013; 1: e26314.
    1. Rothwell PE, Fuccillo MV, Maxeiner S, Hayton SJ, Gokce O, Lim BK, et al.Autism-associated neuroligin-3 mutations commonly impair striatal circuits to boost repetitive behaviors. Cell 2014; 158: 198–212.
    1. Ryan MD, King AM, Thomas GP. Cleavage of foot-and-mouth disease virus polyprotein is mediated by residues located within a 19 amino acid sequence. J Gen Virol 1991; 72 (Pt 11): 2727–32.
    1. Saitsu H, Kato M, Mizuguchi T, Hamada K, Osaka H, Tohyama J, et novo mutations in the gene encoding STXBP1 (MUNC18-1) cause early infantile epileptic encephalopathy. Nat Genet 2008; 40: 782–8.
    1. Saitsu H, Kato M, Okada I, Orii KE, Higuchi T, Hoshino H, et al.STXBP1 mutations in early infantile epileptic encephalopathy with suppression-burst pattern. Epilepsia 2010; 51: 2397–405.
    1. Santos TC, Wierda K, Broeke JH, Toonen RF, Verhage M. Early golgi abnormalities and neurodegeneration upon loss of presynaptic proteins Munc18-1, Syntaxin-1, or SNAP-25. J Neurosci 2017; 37: 4525–39.
    1. Schmitz SK, Hjorth JJ, Joemai RM, Wijntjes R, Eijgenraam S, de Bruijn P, et al.Automated analysis of neuronal morphology, synapse number and synaptic recruitment. J Neurosci Methods 2011; 195: 185–93.
    1. Schmitz SK, King C, Kortleven C, Huson V, Kroon T, Kevenaar JT, et al.Presynaptic inhibition upon CB1 or mGlu2/3 receptor activation requires ERK/MAPK phosphorylation of Munc18-1. EMBO J 2016; 35: 1236–50.
    1. Schubert J, Siekierska A, Langlois M, May P, Huneau C, Becker F, et al.Mutations in STX1B, encoding a presynaptic protein, cause fever-associated epilepsy syndromes. Nat Genet 2014; 46: 1327–32.
    1. Stamberger H, Nikanorova M, Willemsen MH, Accorsi P, Angriman M, Baier H, et al.STXBP1 encephalopathy: a neurodevelopmental disorder including epilepsy. Neurology 2016; 86: 954–62.
    1. Toonen RF, Wierda K, Sons MS, de Wit H, Cornelisse LN, Brussaard A, et al.Munc18-1 expression levels control synapse recovery by regulating readily releasable pool size. Proc Natl Acad Sci USA 2006; 103: 18332–7.
    1. Toonen RF, de Vries KJ, Zalm R, Südhof TC, Verhage M. Munc18-1 stabilizes syntaxin 1, but is not essential for syntaxin 1 targeting and SNARE complex formation. J Neurochem 2005; 93: 1393–400.
    1. Toonen RF, Verhage M. Munc18-1 in secretion: lonely Munc joins SNARE team and takes control. Trends Neurosci 2007; 30: 564–72.
    1. Vanden Berghe T, Hulpiau P, Martens L, Vandenbroucke RE, Van Wonterghem E, Perry SW, et al.Passenger mutations confound interpretation of all genetically modified congenic mice. Immunity 2015; 43: 200–9.
    1. Vatta M, Tennison MB, Aylsworth AS, Turcott CM, Guerra MP, Eng CM, et al.A novel STXBP1 mutation causes focal seizures with neonatal onset. J Child Neurol 2012; 27: 811–4.
    1. Verhage M, Maia AS, Plomp JJ, Brussaard AB, Heeroma JH, Vermeer H, et al.Synaptic assembly of the brain in the absence of neurotransmitter secretion. Science 2000; 287: 864–9.
    1. Verhoeven KJF, Simonsen KL, McIntyre LM. Implementing false discovery rate control: increasing your power. OIKOS 2005; 108: 643–47.
    1. Voets T, Toonen RF, Brian EC, de Wit H, Moser T, Rettig J, et al.Munc18-1 promotes large dense-core vesicle docking. Neuron 2001; 31: 581–91.
    1. Wagnon JL, Korn MJ, Parent R, Tarpey TA, Jones JM, Hammer MF, et al.Convulsive seizures and SUDEP in a mouse model of SCN8A epileptic encephalopathy. Hum Mol Genet 2015; 24: 506–15.
    1. Watanabe S, Yamamori S, Otsuka S, Saito M, Suzuki E, Kataoka M, et al.Epileptogenesis and epileptic maturation in phosphorylation site-specific SNAP-25 mutant mice. Epilepsy Res 2015; 115: 30–44.
    1. Weckhuysen S, Holmgren P, Hendrickx R, Jansen AC, Hasaerts D, Dielman C, et al.Reduction of seizure frequency after epilepsy surgery in a patient with STXBP1 encephalopathy and clinical description of six novel mutation carriers. Epilepsia 2013; 54: e74–80.
    1. Wierda KD, Toonen RF, de Wit H, Brussaard AB, Verhage M. Interdependence of PKC-dependent and PKC-independent pathways for Presynaptic plasticity. Neuron 2007; 54: 275–90.
    1. Wolfer DP, Crusio WE, Lipp HP. Knockout mice: simple solutions to the problems of genetic background and flanking genes [Review]. Trends Neurosci 2002; 25: 336–40.
    1. Yamamoto T, Shimojima K, Yano T, Ueda Y, Takayama R, Ikeda H, et al.Loss-of-function mutations of STXBP1 in patients with epileptic encephalopathy. Brain Dev 2016; 38: 280–4.
    1. Yamashita S, Chiyonobu T, Yoshida M, Maeda H, Zuiki M, Kidowaki S, et al.Mislocalization of syntaxin-1 and impaired neurite growth observed in a human iPSC model for STXBP1-related epileptic encephalopathy. Epilepsia 2016; 57: e81–6.
    1. Yu FH, Mantegazza M, Westenbroek RE, Robbins CA, Kalume F, Burton KA, et al.Reduced sodium current in GABAergic interneurons in a mouse model of severe myoclonic epilepsy in infancy. Nat Neurosci 2006; 9: 1142–9.

Source: PubMed

3
Předplatit